101
|
Viral Membrane Fusion Proteins and RNA Sorting Mechanisms for the Molecular Delivery by Exosomes. Cells 2021; 10:cells10113043. [PMID: 34831268 PMCID: PMC8622164 DOI: 10.3390/cells10113043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022] Open
Abstract
The advancement of precision medicine critically depends on the robustness and specificity of the carriers used for the targeted delivery of effector molecules in the human body. Numerous nanocarriers have been explored in vivo, to ensure the precise delivery of molecular cargos via tissue-specific targeting, including the endocrine part of the pancreas, thyroid, and adrenal glands. However, even after reaching the target organ, the cargo-carrying vehicle needs to enter the cell and then escape lysosomal destruction. Most artificial nanocarriers suffer from intrinsic limitations that prevent them from completing the specific delivery of the cargo. In this respect, extracellular vesicles (EVs) seem to be the natural tool for payload delivery due to their versatility and low toxicity. However, EV-mediated delivery is not selective and is usually short-ranged. By inserting the viral membrane fusion proteins into exosomes, it is possible to increase the efficiency of membrane recognition and also ease the process of membrane fusion. This review describes the molecular details of the viral-assisted interaction between the target cell and EVs. We also discuss the question of the usability of viral fusion proteins in developing extracellular vesicle-based nanocarriers with a higher efficacy of payload delivery. Finally, this review specifically highlights the role of Gag and RNA binding proteins in RNA sorting into EVs.
Collapse
|
102
|
Mani S, Sevanan M, Krishnamoorthy A, Sekar S. A systematic review of molecular approaches that link mitochondrial dysfunction and neuroinflammation in Parkinson's disease. Neurol Sci 2021; 42:4459-4469. [PMID: 34480241 DOI: 10.1007/s10072-021-05551-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 08/07/2021] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative disorder that affects 1% of the population worldwide. Etiology of PD is likely to be multi-factorial such as protein misfolding, mitochondrial dysfunction, oxidative stress, and neuroinflammation that contributes to the pathology of Parkinson's disease (PD), numerous studies have shown that mitochondrial dysfunction may play a key role in the dopaminergic neuronal loss. In multiple ways, the two most important are the activation of neuroinflammation and mitochondrial dysfunction, while mitochondrial dysfunction could cause neuroinflammation and vice versa. Thus, the mitochondrial proteins are the highly promising target for the development of PD. However, the limited amount of dopaminergic neurons prevented the detailed investigation of Parkinson's disease with regard to mitochondrial dysfunction. Both genetic and environmental factors are also associated with mitochondrial dysfunction and PD pathogenesis. The induction of PD by neurotoxins that inhibit mitochondrial complex I provide direct evidence linking mitochondrial dysfunction to PD. A decrease of mitochondrial complex I activity is observed in PD brain and in neurotoxin- or genetic factor-induced in vitro and in vivo models. Moreover, PINK1, Parkin, DJ-1 and LRRK2 mitochondrial PD gene products have important roles in mitophagy, a cellular process that clear damaged mitochondria. This review paper would discuss the evidence for the mitochondrial dysfunction and neuroinflammation in PD.
Collapse
Affiliation(s)
- Sugumar Mani
- Research and Development Centre, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Murugan Sevanan
- Department of Biotechnology, Karunya Institute of Technology and Sciences, Karunya Nagar, Coimbatore, Tamil Nadu, 641114, India.
| | | | - Sathiya Sekar
- Department of Biotechnology, Dr.M.G.R Educational Research Institute, Chennai, India
| |
Collapse
|
103
|
Key Regulators of Autophagosome Closure. Cells 2021; 10:cells10112814. [PMID: 34831036 PMCID: PMC8616111 DOI: 10.3390/cells10112814] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/29/2021] [Accepted: 10/16/2021] [Indexed: 12/11/2022] Open
Abstract
Autophagy is an evolutionarily conserved pathway, in which cytoplasmic components are sequestered within double-membrane vesicles called autophagosomes and then transported into lysosomes or vacuoles for degradation. Over 40 conserved autophagy-related (ATG) genes define the core machinery for the five processes of autophagy: initiation, nucleation, elongation, closure, and fusion. In this review, we focus on one of the least well-characterized events in autophagy, namely the closure of the isolation membrane/phagophore to form the sealed autophagosome. This process is tightly regulated by ESCRT machinery, ATG proteins, Rab GTPase and Rab-related proteins, SNAREs, sphingomyelin, and calcium. We summarize recent progress in the regulation of autophagosome closure and discuss the key questions remaining to be addressed.
Collapse
|
104
|
The Role of MSCs and Cell Fusion in Tissue Regeneration. Int J Mol Sci 2021; 22:ijms222010980. [PMID: 34681639 PMCID: PMC8535885 DOI: 10.3390/ijms222010980] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023] Open
Abstract
Regenerative medicine is concerned with the investigation of therapeutic agents that can be used to promote the process of regeneration after injury or in different diseases. Mesenchymal stem/stromal cells (MSCs) and their secretome—including extracellular vesicles (EVs) are of great interest, due to their role in tissue regeneration, immunomodulatory capacity and low immunogenicity. So far, clinical studies are not very conclusive as they show conflicting efficacies regarding the use of MSCs. An additional process possibly involved in regeneration might be cell fusion. This process occurs in both a physiological and a pathophysiological context and can be affected by immune response due to inflammation. In this review the role of MSCs and cell fusion in tissue regeneration is discussed.
Collapse
|
105
|
Yin X, Chen L, Yuan S, Liu L, Gao Z. A robust high-throughput fluorescent polarization assay for the evaluation and screening of SARS-CoV-2 fusion inhibitors. Bioorg Chem 2021; 116:105362. [PMID: 34598089 PMCID: PMC8457910 DOI: 10.1016/j.bioorg.2021.105362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is a serious threat to global health. One attractive antiviral target is the membrane fusion mechanism employed by the virus to gain access to the host cell. Here we report a robust protein-based fluorescent polarization assay, that mimicking the formation of the six-helix bundle (6-HB) process during the membrane fusion, for the evaluation and screening of SARS-CoV-2 fusion Inhibitors. The IC50 of known inhibitors, HR2P, EK1, and Salvianolic acid C (Sal-C) were measured to be 6.1 nM, 2.5 nM, and 8.9 µM respectively. In addition, we found Sal-A has a slightly lower IC50 (3.9 µM) than Sal-C. Interestingly, simple caffeic acid can also disrupt the formation of 6-HB with a sub-mM concentration. Pilot high throughput screening (HTS) of a small marine natural product library validates the assay with a Z’ factor close to 0.8. We envision the current assay provides a convenient way to screen SARS-CoV-2 fusion inhibitors and assess their binding affinity.
Collapse
Affiliation(s)
- Xinjian Yin
- School of Marine Science, Sun Yat-sen University, Zhuhai 519080, China
| | - Litong Chen
- School of Marine Science, Sun Yat-sen University, Zhuhai 519080, China
| | - Siwen Yuan
- School of Marine Science, Sun Yat-sen University, Zhuhai 519080, China
| | - Lan Liu
- School of Marine Science, Sun Yat-sen University, Zhuhai 519080, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519080, China
| | - Zhizeng Gao
- School of Marine Science, Sun Yat-sen University, Zhuhai 519080, China; State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519080, China.
| |
Collapse
|
106
|
Tang J, Frascaroli G, Zhou X, Knickmann J, Brune W. Cell Fusion and Syncytium Formation in Betaherpesvirus Infection. Viruses 2021; 13:v13101973. [PMID: 34696402 PMCID: PMC8537622 DOI: 10.3390/v13101973] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022] Open
Abstract
Cell–cell fusion is a fundamental and complex process that occurs during reproduction, organ and tissue growth, cancer metastasis, immune response, and infection. All enveloped viruses express one or more proteins that drive the fusion of the viral envelope with cellular membranes. The same proteins can mediate the fusion of the plasma membranes of adjacent cells, leading to the formation of multinucleated syncytia. While cell–cell fusion triggered by alpha- and gammaherpesviruses is well-studied, much less is known about the fusogenic potential of betaherpesviruses such as human cytomegalovirus (HCMV) and human herpesviruses 6 and 7 (HHV-6 and HHV-7). These are slow-growing viruses that are highly prevalent in the human population and associated with several diseases, particularly in individuals with an immature or impaired immune system such as fetuses and transplant recipients. While HHV-6 and HHV-7 are strictly lymphotropic, HCMV infects a very broad range of cell types including epithelial, endothelial, mesenchymal, and myeloid cells. Syncytia have been observed occasionally for all three betaherpesviruses, both during in vitro and in vivo infection. Since cell–cell fusion may allow efficient spread to neighboring cells without exposure to neutralizing antibodies and other host immune factors, viral-induced syncytia may be important for viral dissemination, long-term persistence, and pathogenicity. In this review, we provide an overview of the viral and cellular factors and mechanisms identified so far in the process of cell–cell fusion induced by betaherpesviruses and discuss the possible consequences for cellular dysfunction and pathogenesis.
Collapse
Affiliation(s)
- Jiajia Tang
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; (J.T.); (G.F.); (X.Z.); (J.K.)
- Center for Single-Cell Omics, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Giada Frascaroli
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; (J.T.); (G.F.); (X.Z.); (J.K.)
| | - Xuan Zhou
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; (J.T.); (G.F.); (X.Z.); (J.K.)
| | - Jan Knickmann
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; (J.T.); (G.F.); (X.Z.); (J.K.)
| | - Wolfram Brune
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; (J.T.); (G.F.); (X.Z.); (J.K.)
- Correspondence:
| |
Collapse
|
107
|
Liu H, Kumar R, Zhong C, Gorji S, Paniushkina L, Masood R, Wittel UA, Fuchs H, Nazarenko I, Hirtz M. Rapid Capture of Cancer Extracellular Vesicles by Lipid Patch Microarrays. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2008493. [PMID: 34309083 PMCID: PMC11468818 DOI: 10.1002/adma.202008493] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/12/2021] [Indexed: 06/13/2023]
Abstract
Extracellular vesicles (EVs) contain various bioactive molecules such as DNA, RNA, and proteins, and play a key role in the regulation of cancer progression. Furthermore, cancer-associated EVs carry specific biomarkers and can be used in liquid biopsy for cancer detection. However, it is still technically challenging and time consuming to detect or isolate cancer-associated EVs from complex biofluids (e.g., blood). Here, a novel EV-capture strategy based on dip-pen nanolithography generated microarrays of supported lipid membranes is presented. These arrays carry specific antibodies recognizing EV- and cancer-specific surface biomarkers, enabling highly selective and efficient capture. Importantly, it is shown that the nucleic acid cargo of captured EVs is retained on the lipid array, providing the potential for downstream analysis. Finally, the feasibility of EV capture from patient sera is demonstrated. The demonstrated platform offers rapid capture, high specificity, and sensitivity, with only a small need in analyte volume and without additional purification steps. The platform is applied in context of cancer-associated EVs, but it can easily be adapted to other diagnostic EV targets by use of corresponding antibodies.
Collapse
Affiliation(s)
- Hui‐Yu Liu
- Institute of Nanotechnology (INT) & Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT)Hermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Ravi Kumar
- Institute of Nanotechnology (INT) & Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT)Hermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Chunting Zhong
- Institute of Nanotechnology (INT) & Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT)Hermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Saleh Gorji
- Institute of Nanotechnology (INT) & Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT)Hermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
- Joint Research Laboratory Nanomaterials (KIT and TUD) at Technische Universität Darmstadt (TUD)Jovanka‐Bontschits‐Str. 264287DarmstadtGermany
| | - Liliia Paniushkina
- Institute for Infection Prevention and Hospital EpidemiologyMedical CentreFaculty of MedicineUniversity of FreiburgBreisacher Straße 115 B79106FreiburgGermany
| | - Ramsha Masood
- Institute for Infection Prevention and Hospital EpidemiologyMedical CentreFaculty of MedicineUniversity of FreiburgBreisacher Straße 115 B79106FreiburgGermany
| | - Uwe A. Wittel
- Department of General and Visceral SurgeryCentre of SurgeryMedical CentreFaculty of MedicineUniversity of FreiburgBreisacher Str. 8679110FreiburgGermany
| | - Harald Fuchs
- Institute of Nanotechnology (INT) & Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT)Hermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
- Physikalisches Institut & Center for Nanotechnology (CeNTech)Westfälische Wilhelms‐UniversitätWilhelm‐Klemm‐Straße 1048149MünsterGermany
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital EpidemiologyMedical CentreFaculty of MedicineUniversity of FreiburgBreisacher Straße 115 B79106FreiburgGermany
- German Cancer Consortium (DKTK)Partner Site Freiburg and German Cancer Research Center (DKFZ)Im Neuenheimer Feld 28069120HeidelbergGermany
| | - Michael Hirtz
- Institute of Nanotechnology (INT) & Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT)Hermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| |
Collapse
|
108
|
Conventional Molecular and Novel Structural Mechanistic Insights into Orderly Organelle Interactions. Chem Res Chin Univ 2021. [DOI: 10.1007/s40242-021-1191-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
109
|
Taheri M, Bahrami A, Habibi P, Nouri F. A Review on the Serum Electrolytes and Trace Elements Role in the Pathophysiology of COVID-19. Biol Trace Elem Res 2021; 199:2475-2481. [PMID: 32901413 PMCID: PMC7478435 DOI: 10.1007/s12011-020-02377-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/02/2020] [Indexed: 01/01/2023]
Abstract
All the world is involved in the COVID-19 disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Coronavirus is a positive-sense RNA and has an envelope. There is no specific drug for this disease and treatment methods are limited. Malnutrition and electrolyte imbalance can make dysfunction in the immune system and impairment of the immune system causes increasing the risk of infection. Understanding the aspects of biological features of the virus will help the development of diagnostic tests, pharmacological therapies, and vaccines. Here, we review and discuss increasing and decreasing some trace elements and imbalance of serum and plasma electrolytes involving in COVID-19.
Collapse
Affiliation(s)
- Mohammad Taheri
- Department of Medical Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Bahrami
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Parisa Habibi
- Neurophysiology Research Center, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Nouri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
110
|
Wu Z, Dharan N, McDargh ZA, Thiyagarajan S, O'Shaughnessy B, Karatekin E. The neuronal calcium sensor Synaptotagmin-1 and SNARE proteins cooperate to dilate fusion pores. eLife 2021; 10:68215. [PMID: 34190041 PMCID: PMC8294851 DOI: 10.7554/elife.68215] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
All membrane fusion reactions proceed through an initial fusion pore, including calcium-triggered release of neurotransmitters and hormones. Expansion of this small pore to release cargo is energetically costly and regulated by cells, but the mechanisms are poorly understood. Here, we show that the neuronal/exocytic calcium sensor Synaptotagmin-1 (Syt1) promotes expansion of fusion pores induced by SNARE proteins. Pore dilation relied on calcium-induced insertion of the tandem C2 domain hydrophobic loops of Syt1 into the membrane, previously shown to reorient the C2 domain. Mathematical modelling suggests that C2B reorientation rotates a bound SNARE complex so that it exerts force on the membranes in a mechanical lever action that increases the height of the fusion pore, provoking pore dilation to offset the bending energy penalty. We conclude that Syt1 exerts novel non-local calcium-dependent mechanical forces on fusion pores that dilate pores and assist neurotransmitter and hormone release.
Collapse
Affiliation(s)
- Zhenyong Wu
- Department of Cellular and Molecular Physiology, Yale University, New Haven, United States.,Nanobiology Institute, Yale University, West Haven, United States
| | - Nadiv Dharan
- Department of Chemical Engineering, Columbia University, New York, United States
| | - Zachary A McDargh
- Department of Chemical Engineering, Columbia University, New York, United States
| | - Sathish Thiyagarajan
- Department of Chemical Engineering, Columbia University, New York, United States
| | - Ben O'Shaughnessy
- Department of Chemical Engineering, Columbia University, New York, United States
| | - Erdem Karatekin
- Department of Cellular and Molecular Physiology, Yale University, New Haven, United States.,Nanobiology Institute, Yale University, West Haven, United States.,Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, United States.,Saints-Pères Paris Institute for the Neurosciences (SPPIN), Université de Paris, Centre National de la Recherche Scientifique (CNRS) UMR 8003, Paris, France
| |
Collapse
|
111
|
Krag TO, Holm-Yildiz S, Witting N, Vissing J. Autophagy is affected in patients with hypokalemic periodic paralysis: an involvement in vacuolar myopathy? Acta Neuropathol Commun 2021; 9:109. [PMID: 34120654 PMCID: PMC8201813 DOI: 10.1186/s40478-021-01212-8] [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: 04/07/2021] [Accepted: 06/07/2021] [Indexed: 11/18/2022] Open
Abstract
Hypokalemic periodic paralysis is an autosomal dominant, rare disorder caused by variants in the genes for voltage-gated calcium channel CaV1.1 (CACNA1S) and NaV1.4 (SCN4A). Patients with hypokalemic periodic paralysis may suffer from periodic paralysis alone, periodic paralysis co-existing with permanent weakness or permanent weakness alone. Hypokalemic periodic paralysis has been known to be associated with vacuolar myopathy for decades, and that vacuoles are a universal feature regardless of phenotype. Hence, we wanted to investigate the nature and cause of the vacuoles. Fourteen patients with the p.R528H variation in the CACNA1S gene was included in the study. Histology, immunohistochemistry and transmission electron microscopy was used to assess general histopathology, ultrastructure and pattern of expression of proteins related to muscle fibres and autophagy. Western blotting and real-time PCR was used to determine the expression levels of proteins and mRNA of the proteins investigated in immunohistochemistry. Histology and transmission electron microscopy revealed heterogenous vacuoles containing glycogen, fibrils and autophagosomes. Immunohistochemistry demonstrated autophagosomes and endosomes arrested at the pre-lysosome fusion stage. Expression analysis showed a significant decrease in levels of proteins an mRNA involved in autophagy in patients, suggesting a systemic effect. However, activation level of the master regulator of autophagy gene transcription, TFEB, did not differ between patients and controls, suggesting competing control over autophagy gene transcription by nutritional status and calcium concentration, both controlling TFEB activity. The findings suggest that patients with hypokalemic periodic paralysis have disrupted autophagic processing that contribute to the vacuoles seen in these patients.
Collapse
|
112
|
Elson A, Stein M, Rabie G, Barnea-Zohar M, Winograd-Katz S, Reuven N, Shalev M, Sekeres J, Kanaan M, Tuckermann J, Geiger B. Sorting Nexin 10 as a Key Regulator of Membrane Trafficking in Bone-Resorbing Osteoclasts: Lessons Learned From Osteopetrosis. Front Cell Dev Biol 2021; 9:671210. [PMID: 34095139 PMCID: PMC8173195 DOI: 10.3389/fcell.2021.671210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/23/2021] [Indexed: 12/30/2022] Open
Abstract
Bone homeostasis is a complex, multi-step process, which is based primarily on a tightly orchestrated interplay between bone formation and bone resorption that is executed by osteoblasts and osteoclasts (OCLs), respectively. The essential physiological balance between these cells is maintained and controlled at multiple levels, ranging from regulated gene expression to endocrine signals, yet the underlying cellular and molecular mechanisms are still poorly understood. One approach for deciphering the mechanisms that regulate bone homeostasis is the characterization of relevant pathological states in which this balance is disturbed. In this article we describe one such “error of nature,” namely the development of acute recessive osteopetrosis (ARO) in humans that is caused by mutations in sorting nexin 10 (SNX10) that affect OCL functioning. We hypothesize here that, by virtue of its specific roles in vesicular trafficking, SNX10 serves as a key selective regulator of the composition of diverse membrane compartments in OCLs, thereby affecting critical processes in the sequence of events that link the plasma membrane with formation of the ruffled border and with extracellular acidification. As a result, SNX10 determines multiple features of these cells either directly or, as in regulation of cell-cell fusion, indirectly. This hypothesis is further supported by the similarities between the cellular defects observed in OCLs form various models of ARO, induced by mutations in SNX10 and in other genes, which suggest that mutations in the known ARO-associated genes act by disrupting the same plasma membrane-to-ruffled border axis, albeit to different degrees. In this article, we describe the population genetics and spread of the original arginine-to-glutamine mutation at position 51 (R51Q) in SNX10 in the Palestinian community. We further review recent studies, conducted in animal and cellular model systems, that highlight the essential roles of SNX10 in critical membrane functions in OCLs, and discuss possible future research directions that are needed for challenging or substantiating our hypothesis.
Collapse
Affiliation(s)
- Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Merle Stein
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Grace Rabie
- Hereditary Research Laboratory and Department of Life Sciences, Bethlehem University, Bethlehem, Palestine
| | - Maayan Barnea-Zohar
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | | | - Nina Reuven
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Moran Shalev
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Juraj Sekeres
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Moien Kanaan
- Hereditary Research Laboratory and Department of Life Sciences, Bethlehem University, Bethlehem, Palestine
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Benjamin Geiger
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
113
|
Malfunctioning CD106-positive, short-term hematopoietic stem cells trigger diabetic neuropathy in mice by cell fusion. Commun Biol 2021; 4:575. [PMID: 33990693 PMCID: PMC8121918 DOI: 10.1038/s42003-021-02082-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 03/25/2021] [Indexed: 12/16/2022] Open
Abstract
Diabetic neuropathy is an incurable disease. We previously identified a mechanism by which aberrant bone marrow-derived cells (BMDCs) pathologically expressing proinsulin/TNF-α fuse with residential neurons to impair neuronal function. Here, we show that CD106-positive cells represent a significant fraction of short-term hematopoietic stem cells (ST-HSCs) that contribute to the development of diabetic neuropathy in mice. The important role for these cells is supported by the fact that transplantation of either whole HSCs or CD106-positive ST-HSCs from diabetic mice to non-diabetic mice produces diabetic neuronal dysfunction in the recipient mice via cell fusion. Furthermore, we show that transient episodic hyperglycemia produced by glucose injections leads to abnormal fusion of pathological ST-HSCs with residential neurons, reproducing neuropathy in nondiabetic mice. In conclusion, we have identified hyperglycemia-induced aberrant CD106-positive ST-HSCs underlie the development of diabetic neuropathy. Aberrant CD106-positive ST-HSCs constitute a novel therapeutic target for the treatment of diabetic neuropathy. Katagi et al. show that abnormal bone marrow-derived cells originated from hematopoietic stem cells (CD106-positive short-term HSCs) aberrantly fuse with neurons to develop diabetic neuropathy. This study suggests that the pathological abnormality is memorized in the bone marrow and that it cannot be erased by conventional therapy.
Collapse
|
114
|
Study of the solubilisation process of bacterial model membranes induced by DDAO. EUROPEAN PHARMACEUTICAL JOURNAL 2021. [DOI: 10.2478/afpuc-2020-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Solubilisation of two bacterial model membranes induced by N,N-dimethyl-1-dodecanamine-N-oxide (DDAO) was studied. The first model membrane consisted of a mixture of palmitoyloleoylphosphatidylethanolamine (POPE) and palmitoyloleoylphosphatidylglycerol (POPG) in a molar ratio 0.6:0.4 mol/mol, and a second model membrane was enriched with tetraoleoylcardiolipin (TOCL) with a composition POPE-POPG-TOCL = 0.67:0.23:0.1 mol/mol/mol. Solubilisation of these model membranes was studied by static light scattering (nephelometry). Effective ratio Re (the amount of DDAO integrated into the bilayer to the amount of lipid) at different steps of the solubilisation process was determined. The molar partition coefficient of DDAO was calculated – in case of the POPE-POPG membrane, Kp = 5,300 ± 400, for the POPE-POPG-TOCL membrane, Kp = 6,500 ± 500.
Collapse
|
115
|
Barnea-Zohar M, Winograd-Katz SE, Shalev M, Arman E, Reuven N, Roth L, Golani O, Stein M, Thalji F, Kanaan M, Tuckermann J, Geiger B, Elson A. An SNX10-dependent mechanism downregulates fusion between mature osteoclasts. J Cell Sci 2021; 134:261809. [PMID: 33975343 PMCID: PMC8182410 DOI: 10.1242/jcs.254979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 03/19/2021] [Indexed: 01/13/2023] Open
Abstract
Homozygosity for the R51Q mutation in sorting nexin 10 (SNX10) inactivates osteoclasts (OCLs) and induces autosomal recessive osteopetrosis in humans and in mice. We show here that the fusion of wild-type murine monocytes to form OCLs is highly regulated, and that its extent is limited by blocking fusion between mature OCLs. In contrast, monocytes from homozygous R51Q SNX10 mice fuse uncontrollably, forming giant dysfunctional OCLs that can become 10- to 100-fold larger than their wild-type counterparts. Furthermore, mutant OCLs display reduced endocytotic activity, suggesting that their deregulated fusion is due to alterations in membrane homeostasis caused by loss of SNX10 function. This is supported by the finding that the R51Q SNX10 protein is unstable and exhibits altered lipid-binding properties, and is consistent with a key role for SNX10 in vesicular trafficking. We propose that OCL size and functionality are regulated by a cell-autonomous SNX10-dependent mechanism that downregulates fusion between mature OCLs. The R51Q mutation abolishes this regulatory activity, leading to excessive fusion, loss of bone resorption capacity and, consequently, to an osteopetrotic phenotype in vivo. This article has an associated First Person interview with the joint first authors of the paper. Summary: Fusion of monocytes to become bone-resorbing osteoclasts is limited by an SNX10-dependent cell-autonomous mechanism. Loss of SNX10 function deregulates fusion and generates giant inactive osteoclasts.
Collapse
Affiliation(s)
- Maayan Barnea-Zohar
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Moran Shalev
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Esther Arman
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nina Reuven
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Lee Roth
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ofra Golani
- Department of Life Sciences Core Facilities, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Merle Stein
- Department of Biology, Institute of Comparative Molecular Endocrinology, University of Ulm, 89081 Ulm, Germany
| | - Fadi Thalji
- Department of Orthopedics, Istishari Arab Hospital, Ramallah, Palestine
| | - Moien Kanaan
- Hereditary Research Laboratory and Department of Life Sciences, Bethlehem University, Bethlehem 0045866, Palestine
| | - Jan Tuckermann
- Department of Biology, Institute of Comparative Molecular Endocrinology, University of Ulm, 89081 Ulm, Germany
| | - Benjamin Geiger
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
116
|
Xu H, Cai M, Gao J, Shi Y, Chen J, Wu Q, Zhang J, Jiang J, Wang H. Membrane protein density determining membrane fusion revealed by dynamic fluorescence imaging. Talanta 2021; 226:122091. [PMID: 33676648 DOI: 10.1016/j.talanta.2021.122091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 01/04/2023]
Abstract
Membrane fusion is fundamental to biological activity of cells, so disclosingits relevant mechanism is very important for understanding various cell functions. Although artificial model systems have been developed to uncover the mechanism of membrane fusion, key factors determining the mode of membrane fusion remain unclear. Based on the construction of different types of liposome vesicles, we used a dynamic fluorescence imaging method to investigate the effect of membrane protein distribution density on membrane fusion. Time-resolved imaging revealed that protein-free pure phospholipid vesicles themselves occurred full membrane fusion. Moreover, we prepared proteoliposomes with increasing protein-to-lipid ratio to better reflect the characteristic of membrane structure in vivo. Our data showed that pure phospholipid vesicles no longer fused with the proteoliposomes that in a higher protein proportion, indicating dense membrane proteins may hinder membrane fusion. A further comparative analysis of the interactions of pure phospholipid vesicles with the cell membrane / giant plasma membrane vesicles (GPMVs) / protein-free giant unilamellar vesicles (GUVs) confirmed the inhibitory effect of dense membrane proteins on membrane fusion. Our work demonstrates the membrane protein density influences the mode of membrane fusion and lays a foundation for constructing quasi-native membrane fusion models in vitro.
Collapse
Affiliation(s)
- Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China; Graduate University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China
| | - Yan Shi
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China
| | - Junling Chen
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, PR China
| | - Qiang Wu
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, PR China
| | - Jinrui Zhang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China
| | - Junguang Jiang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China; University of Science and Technology of China, Hefei, Anhui, 230026, PR China; Laboratory for Marine Biology and Biotechnology, Qing Dao National Laboratory for Marine Science and Technology, Qingdao, Shandong, 266237, PR China.
| |
Collapse
|
117
|
Berg Klenow M, Iversen C, Wendelboe Lund F, Mularski A, Busk Heitmann AS, Dias C, Nylandsted J, Simonsen AC. Annexins A1 and A2 Accumulate and Are Immobilized at Cross-Linked Membrane-Membrane Interfaces. Biochemistry 2021; 60:1248-1259. [PMID: 33861586 DOI: 10.1021/acs.biochem.1c00126] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Rapid membrane repair is required to ensure cell survival after rupture of the plasma membrane. The annexin family of proteins is involved in plasma membrane repair (PMR) and is activated by the influx of Ca2+ from the extracellular medium at the site of injury. Annexins A1 and A2 (ANXA1 and ANXA2, respectively) are structurally similar and bind to negatively charged phosphatidylserine (PS) to induce membrane cross-linking and to promote fusion, which are both essential processes that occur during membrane repair. The degree of annexin accumulation and the annexin mobility at cross-linked membranes are important aspects of ANXA1 and ANXA2 function in repair. Here, we quantify ANXA1- and ANXA2-induced membrane cross-linking between giant unilamellar vesicles (GUVs). Time-lapse measurements show that ANXA1 and ANXA2 can induce membrane cross-linking on a time scale compatible with PMR. Cross-linked membrane-membrane interfaces between the GUVs persist in time without fusion, and quantification of confocal microscopy images demonstrates that ANXA1, ANXA2, and, to a lesser extent, PS lipids accumulate at the double membrane interface. Fluorescence recovery after photobleaching shows that the annexins are fully immobilized at the double membrane interface, whereas PS lipids display a 75% decrease in mobility. In addition, the complete immobilization of annexins between two membranes indicates a high degree of network formation between annexins, suggesting that membrane cross-linking is mainly driven by protein-protein interactions.
Collapse
Affiliation(s)
- Martin Berg Klenow
- Department of Physics, Chemistry and Pharmacy (FKF), University of Southern Denmark (SDU), Campusvej 55, 5230 Odense M, Denmark
| | - Christoffer Iversen
- Department of Physics, Chemistry and Pharmacy (FKF), University of Southern Denmark (SDU), Campusvej 55, 5230 Odense M, Denmark
| | - Frederik Wendelboe Lund
- Department of Physics, Chemistry and Pharmacy (FKF), University of Southern Denmark (SDU), Campusvej 55, 5230 Odense M, Denmark
| | - Anna Mularski
- Department of Physics, Chemistry and Pharmacy (FKF), University of Southern Denmark (SDU), Campusvej 55, 5230 Odense M, Denmark
| | - Anne Sofie Busk Heitmann
- Membrane Integrity, Danish Cancer Society Research Center, Strandboulevarden 49, 2100 Copenhagen Ø, Denmark
| | - Catarina Dias
- Membrane Integrity, Danish Cancer Society Research Center, Strandboulevarden 49, 2100 Copenhagen Ø, Denmark
| | - Jesper Nylandsted
- Membrane Integrity, Danish Cancer Society Research Center, Strandboulevarden 49, 2100 Copenhagen Ø, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | - Adam Cohen Simonsen
- Department of Physics, Chemistry and Pharmacy (FKF), University of Southern Denmark (SDU), Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
118
|
Pattnaik GP, Chakraborty H. Fusogenic Effect of Cholesterol Prevails over the Inhibitory Effect of a Peptide-Based Membrane Fusion Inhibitor. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:3477-3489. [PMID: 33689373 DOI: 10.1021/acs.langmuir.1c00319] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Membrane fusion is the primary step in the entry of enveloped viruses into the host cell. Membrane composition modulates the membrane fusion by changing the organization dynamics of the fusion proteins, peptides, and membranes. The asymmetric lipid compositions of the viral envelope and the host cell influence the membrane fusion. Cholesterol is an important constituent of mammalian cells and plays a vital role in the entry of several viruses. In our pursuit of developing peptide-based general fusion inhibitors, we have previously shown that a coronin 1-derived peptide, TG-23, inhibited polyethylene glycol-induced fusion between symmetric membranes without cholesterol. In this work, we have studied the effect of TG-23 on the polyethylene glycol-mediated fusion between 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and 1,2-dioleoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (DOPG) (60/30/10 mol %) and DOPC/DOPE/DOPG/CH (50/30/10/10 mol %) membranes and between DOPC/DOPE/DOPG (60/30/10 mol %) and DOPC/DOPE/DOPG/CH (40/30/10/20 mol %) membranes. Our results demonstrate that the TG-23 peptide inhibited the fusion between membranes containing 0 and 10 mol % cholesterol though the efficacy is less than that of symmetric fusion between membranes devoid of cholesterol, and the inhibitory efficacy becomes negligible in the fusion between membranes containing 0 and 20 mol % cholesterol. Several steady-state and time-resolved fluorescence spectroscopic techniques have been successfully utilized to evaluate the organization, dynamics, and membrane penetration of the TG-23 peptide. Taken together, our results demonstrate that the reduction of the inhibitory effect of TG-23 in asymmetric membrane fusion containing cholesterol of varying concentrations is not due to the altered peptide structure, organization, and dynamics, rather owing to the intrinsic negative curvature-inducing property of cholesterol. Therefore, the membrane composition is an added complexity in the journey of developing peptide-based membrane fusion inhibitors.
Collapse
Affiliation(s)
| | - Hirak Chakraborty
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha 768 019, India
- Centre of Excellence in Natural Products and Therapeutics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768 019, India
| |
Collapse
|
119
|
Tagoe DNA, Drozda AA, Falco JA, Bechtel TJ, Weerapana E, Gubbels MJ. Ferlins and TgDOC2 in Toxoplasma Microneme, Rhoptry and Dense Granule Secretion. Life (Basel) 2021; 11:217. [PMID: 33803212 PMCID: PMC7999867 DOI: 10.3390/life11030217] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 12/25/2022] Open
Abstract
The host cell invasion process of apicomplexan parasites like Toxoplasma gondii is facilitated by sequential exocytosis of the microneme, rhoptry and dense granule organelles. Exocytosis is facilitated by a double C2 domain (DOC2) protein family. This class of C2 domains is derived from an ancestral calcium (Ca2+) binding archetype, although this feature is optional in extant C2 domains. DOC2 domains provide combinatorial power to the C2 domain, which is further enhanced in ferlins that harbor 5-7 C2 domains. Ca2+ conditionally engages the C2 domain with lipids, membranes, and/or proteins to facilitating vesicular trafficking and membrane fusion. The widely conserved T. gondii ferlins 1 (FER1) and 2 (FER2) are responsible for microneme and rhoptry exocytosis, respectively, whereas an unconventional TgDOC2 is essential for microneme exocytosis. The general role of ferlins in endolysosmal pathways is consistent with the repurposed apicomplexan endosomal pathways in lineage specific secretory organelles. Ferlins can facilitate membrane fusion without SNAREs, again pertinent to the Apicomplexa. How temporal raises in Ca2+ combined with spatiotemporally available membrane lipids and post-translational modifications mesh to facilitate sequential exocytosis events is discussed. In addition, new data on cross-talk between secretion events together with the identification of a new microneme protein, MIC21, is presented.
Collapse
Affiliation(s)
- Daniel N A Tagoe
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | - Allison A Drozda
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | - Julia A Falco
- Department of Chemistry, Boston College, Chestnut Hill, MA 02467, USA
| | - Tyler J Bechtel
- Department of Chemistry, Boston College, Chestnut Hill, MA 02467, USA
| | | | - Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| |
Collapse
|
120
|
Zou M, Du Y, Liu R, Zheng Z, Xu J. Nanocarrier-delivered small interfering RNA for chemoresistant ovarian cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 12:e1648. [PMID: 33682310 DOI: 10.1002/wrna.1648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States. Because success in early screening is limited, and most patients with advanced disease develop resistance to multiple treatment modalities, the overall prognosis of ovarian cancer is poor. Despite the revolutionary role of surgery and chemotherapy in curing ovarian cancer, recurrence remains a major challenge in treatment. Thus, improving our understanding of the pathogenesis of ovarian cancer is essential for developing more effective treatments. In this review, we analyze the underlying molecular mechanisms leading to chemotherapy resistance. We discuss the clinical benefits and potential challenges of using nanocarrier-delivered small interfering RNA to treat chemotherapy-resistant ovarian cancer. We aim to elicit collaborative studies on nanocarrier-delivered small interfering RNA to improve the long-term survival rate and quality of life of patients with ovarian cancer. This article is categorized under: RNA Methods > RNA Nanotechnology Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.
Collapse
Affiliation(s)
- Mingyuan Zou
- Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruizhen Liu
- The First People's Hospital of Wu'an, Wu'an, Hebei, China
| | - Zeliang Zheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
121
|
Zhao Z, Qin P, Huang YW. Lysosomal ion channels involved in cellular entry and uncoating of enveloped viruses: Implications for therapeutic strategies against SARS-CoV-2. Cell Calcium 2021; 94:102360. [PMID: 33516131 PMCID: PMC7825922 DOI: 10.1016/j.ceca.2021.102360] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/21/2022]
Abstract
Ion channels are necessary for correct lysosomal function including degradation of cargoes originating from endocytosis. Almost all enveloped viruses, including coronaviruses (CoVs), enter host cells via endocytosis, and do not escape endosomal compartments into the cytoplasm (via fusion with the endolysosomal membrane) unless the virus-encoded envelope proteins are cleaved by lysosomal proteases. With the ongoing outbreak of severe acute respiratory syndrome (SARS)-CoV-2, endolysosomal two-pore channels represent an exciting and emerging target for antiviral therapies. This review focuses on the latest knowledge of the effects of lysosomal ion channels on the cellular entry and uncoating of enveloped viruses, which may aid in development of novel therapies against emerging infectious diseases such as SARS-CoV-2.
Collapse
Affiliation(s)
- Zhuangzhuang Zhao
- Key Laboratory of Animal Virology of Ministry of Agriculture, Institute of Preventive Veterinary Medicine, Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Pan Qin
- Key Laboratory of Animal Virology of Ministry of Agriculture, Institute of Preventive Veterinary Medicine, Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yao-Wei Huang
- Key Laboratory of Animal Virology of Ministry of Agriculture, Institute of Preventive Veterinary Medicine, Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
122
|
Giuliani AL, Sarti AC, Di Virgilio F. Ectonucleotidases in Acute and Chronic Inflammation. Front Pharmacol 2021; 11:619458. [PMID: 33613285 PMCID: PMC7887318 DOI: 10.3389/fphar.2020.619458] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Ectonucleotidases are extracellular enzymes with a pivotal role in inflammation that hydrolyse extracellular purine and pyrimidine nucleotides, e.g., ATP, UTP, ADP, UDP, AMP and NAD+. Ectonucleotidases, expressed by virtually all cell types, immune cells included, either as plasma membrane-associated or secreted enzymes, are classified into four main families: 1) nucleoside triphosphate diphosphohydrolases (NTPDases), 2) nicotinamide adenine dinucleotide glycohydrolase (NAD glycohydrolase/ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1), 3) ecto-5′-nucleotidase (NT5E), and 4) ecto-nucleotide pyrophosphatase/phosphodiesterases (NPPs). Concentration of ATP, UTP and NAD+ can be increased in the extracellular space thanks to un-regulated, e.g., cell damage or cell death, or regulated processes. Regulated processes include secretory exocytosis, connexin or pannexin hemichannels, ATP binding cassette (ABC) transporters, calcium homeostasis modulator (CALMH) channels, the ATP-gated P2X7 receptor, maxi-anion channels (MACs) and volume regulated ion channels (VRACs). Hydrolysis of extracellular purine nucleotides generates adenosine, an important immunosuppressant. Extracellular nucleotides and nucleosides initiate or dampen inflammation via P2 and P1 receptors, respectively. All these agents, depending on their level of expression or activation and on the agonist concentration, are potent modulators of inflammation and key promoters of host defences, immune cells activation, pathogen clearance, tissue repair and regeneration. Thus, their knowledge is of great importance for a full understanding of the pathophysiology of acute and chronic inflammatory diseases. A selection of these pathologies will be briefly discussed here.
Collapse
Affiliation(s)
- Anna Lisa Giuliani
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Alba Clara Sarti
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesco Di Virgilio
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
123
|
Praharaj PP, Patro BS, Bhutia SK. Dysregulation of mitophagy and mitochondrial homeostasis in cancer stem cells: Novel mechanism for anti-cancer stem cell-targeted cancer therapy. Br J Pharmacol 2021; 179:5015-5035. [PMID: 33527371 DOI: 10.1111/bph.15401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
Despite the potential of cancer medicine, cancer stem cells (CSCs) associated with chemoresistance and disease recurrence are the significant challenges currently opposing the efficacy of available cancer treatment options. Mitochondrial dynamics involving the fission-fusion cycle and mitophagy are the major contributing factors to better adaptation, enabling CSCs to survive and grow better under tumour micro-environment-associated stress. Moreover, mitophagy is balanced with mitochondrial biogenesis to maintain mitochondrial homeostasis in CSCs, which are necessary for the growth and maintenance of CSCs and regulate metabolic switching from glycolysis to oxidative phosphorylation. In this review, we discuss different aspects of mitochondrial dynamics, mitophagy, and mitochondrial homeostasis and their effects on modulating CSCs behaviour during cancer development. Moreover, the efficacy of pharmacological targeting of these cellular processes using anti-CSC drugs in combination with currently available chemotherapeutic drugs improves the patient's survival of aggressive cancer types.
Collapse
Affiliation(s)
- Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India
| | | | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India
| |
Collapse
|
124
|
Löffler PMG, Rabe A, Vogel S. Lipid-Modified Peptide Nucleic Acids: Synthesis and Application to Programmable Liposome Fusion. Methods Mol Biol 2021; 2105:75-96. [PMID: 32088865 DOI: 10.1007/978-1-0716-0243-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Peptide nucleic acids (PNAs) can be modified with aliphatic lipid chains and designed to be water soluble and able to spontaneously insert into phospholipid bilayers. Liposomes with 1.5% negatively charged POPG can be driven to fuse and mix their inner content volumes via functionalization with such lipidated peptide nucleic acids (LiPNAs). During fusion, only low amounts of leakage occur (<5%). We describe here the synthesis and purification of such LiPNAs using an automated peptide synthesizer and the preparation of LiPNA functionalized liposomes. Further, we describe the measurement of LiPNA-induced fusion using a fluorescence-based assay for the content mixing between a liposome population with an encapsulated self-quenching fluorescent dye (SRB) and a buffer-filled liposome population.
Collapse
Affiliation(s)
- Philipp M G Löffler
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Alexander Rabe
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Stefan Vogel
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
125
|
Yang Y, Margam NN. Structural Insights into Membrane Fusion Mediated by Convergent Small Fusogens. Cells 2021; 10:cells10010160. [PMID: 33467484 PMCID: PMC7830690 DOI: 10.3390/cells10010160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/07/2021] [Accepted: 01/13/2021] [Indexed: 12/30/2022] Open
Abstract
From lifeless viral particles to complex multicellular organisms, membrane fusion is inarguably the important fundamental biological phenomena. Sitting at the heart of membrane fusion are protein mediators known as fusogens. Despite the extensive functional and structural characterization of these proteins in recent years, scientists are still grappling with the fundamental mechanisms underlying membrane fusion. From an evolutionary perspective, fusogens follow divergent evolutionary principles in that they are functionally independent and do not share any sequence identity; however, they possess structural similarity, raising the possibility that membrane fusion is mediated by essential motifs ubiquitous to all. In this review, we particularly emphasize structural characteristics of small-molecular-weight fusogens in the hope of uncovering the most fundamental aspects mediating membrane–membrane interactions. By identifying and elucidating fusion-dependent functional domains, this review paves the way for future research exploring novel fusogens in health and disease.
Collapse
|
126
|
Sharma P, Nicoli ER, Serra-Vinardell J, Morimoto M, Toro C, Malicdan MCV, Introne WJ. Chediak-Higashi syndrome: a review of the past, present, and future. ACTA ACUST UNITED AC 2021; 31:31-36. [PMID: 33424983 DOI: 10.1016/j.ddmod.2019.10.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Since the initial description of Chediak-Higashi syndrome (CHS), over 75 years ago, several studies have been conducted to underscore the role of the lysosomal trafficking regulator (LYST) gene in the pathogenesis of disease. CHS is a rare autosomal recessive disorder, which is caused by biallelic mutations in the highly conserved LYST gene. The disease is characterized by partial oculocutaneous albinism, prolonged bleeding, immune and neurologic dysfunction, and risk for the development of hemophagocytic lympohistiocytosis (HLH). The presence of giant secretory granules in leukocytes is the classical diagnostic feature, which distinguishes CHS from closely related Griscelli and Hermansky-Pudlak syndromes. While the exact mechanism of the formation of the giant granules in CHS patients is not understood, dysregulation of LYST function in regulating lysosomal biogenesis has been proposed to play a role. In this review, we discuss the clinical characteristics of the disease and highlight the functional consequences of enlarged lysosomes and lysosome-related organelles (LROs) in CHS.
Collapse
Affiliation(s)
- Prashant Sharma
- Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elena-Raluca Nicoli
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jenny Serra-Vinardell
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marie Morimoto
- Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Camilo Toro
- Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.,Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - May Christine V Malicdan
- Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.,Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.,Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wendy J Introne
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.,Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
127
|
Lu S, Pan X, Chen D, Xie X, Wu Y, Shang W, Jiang X, Sun Y, Fan S, He J. Broad-spectrum antivirals of protoporphyrins inhibit the entry of highly pathogenic emerging viruses. Bioorg Chem 2021; 107:104619. [PMID: 33450541 PMCID: PMC7784547 DOI: 10.1016/j.bioorg.2020.104619] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/18/2020] [Accepted: 12/27/2020] [Indexed: 12/19/2022]
Abstract
PPIX possesses broad antiviral activities in vitro against a panel of enveloped viruses. PPIX interacts with the lipids of enveloped virions, thereby inhibiting the entry of enveloped viruses into host cells. PPIX shows the antiviral effect in vivo by testing mice infected with the influenza A/Puerto Rico/8/34 (H1N1) virus.
Severe emerging and re-emerging viral infections such as Lassa fever, Avian influenza (AI), and COVID-19 caused by SARS-CoV-2 urgently call for new strategies for the development of broad-spectrum antivirals targeting conserved components in the virus life cycle. Viral lipids are essential components, and viral-cell membrane fusion is the required entry step for most unrelated enveloped viruses. In this paper, we identified a porphyrin derivative of protoporphyrin IX (PPIX) that showed broad antiviral activities in vitro against a panel of enveloped pathogenic viruses including Lassa virus (LASV), Machupo virus (MACV), and SARS-CoV-2 as well as various subtypes of influenza A viral strains with IC50 values ranging from 0.91 ± 0.25 μM to 1.88 ± 0.34 μM. A mechanistic study using influenza A/Puerto Rico/8/34 (H1N1) as a testing strain showed that PPIX inhibits the infection in the early stage of virus entry through biophysically interacting with the hydrophobic lipids of enveloped virions, thereby inhibiting the entry of enveloped viruses into host cells. In addition, the preliminary antiviral activities of PPIX were further assessed by testing mice infected with the influenza A/Puerto Rico/8/34 (H1N1) virus. The results showed that compared with the control group without drug treatment, the survival rate and mean survival time of the mice treated with PPIX were apparently prolonged. These data encourage us to conduct further investigations using PPIX as a lead compound for the rational design of lipid-targeting antivirals for the treatment of infection with enveloped viruses.
Collapse
Affiliation(s)
- Shengsheng Lu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Xiaoyan Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Daiwei Chen
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Xi Xie
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Weijuan Shang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xiaming Jiang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Yuan Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Sheng Fan
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Jian He
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China.
| |
Collapse
|
128
|
Lim JH, Jung HY, Choi JY, Park SH, Kim CD, Kim YL, Cho JH. Hypertension and Electrolyte Disorders in Patients with COVID-19. Electrolyte Blood Press 2020; 18:23-30. [PMID: 33408744 PMCID: PMC7781764 DOI: 10.5049/ebp.2020.18.2.23] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023] Open
Abstract
The worldwide coronavirus disease 2019 (COVID-19) pandemic is still in progress, but much remains unknown about the disease. In this article, we review the association of hypertension or the renin-angiotensin system (RAS) with COVID-19 and the correlation between electrolyte disorders and disease severity. Underlying hypertension is likely to be associated with severe or critical COVID-19, but the relationship is not clear owing to confounding factors. Angiotensin-converting enzyme 2 (ACE2) plays an important role in the non-classical RAS pathway and binds to a receptor binding domain of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The RAS blockade is known to increase ACE2 levels, but controversy remains regarding the effect of RAS blockade therapy in the course of COVID-19. Some reports have indicated a protective effect of RAS blockade on COVID-19, whereas others have reported an association of RAS blockade therapy with the occurrence of severe complications such as acute kidney injury and admission to the intensive care unit. Electrolyte disorders are not uncommon in patients with COVID-19, and severe COVID-19 has frequently shown hypokalemia, hyponatremia, and hypocalcemia. Electrolyte imbalances are caused by alteration of RAS, gastrointestinal loss, effects of proinflammatory cytokines, and renal tubular dysfunction by the invasion of SARS-CoV-2.
Collapse
Affiliation(s)
- Jeong-Hoon Lim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Hee-Yeon Jung
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Ji-Young Choi
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Sun-Hee Park
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Chan-Duck Kim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Yong-Lim Kim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Jang-Hee Cho
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
129
|
Gwanyanya A, Godsmark CN, Kelly-Laubscher R. Ethanolamine: A Potential Promoiety with Additional Effects in the Brain. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 21:108-117. [PMID: 33319663 DOI: 10.2174/1871527319999201211204645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/14/2020] [Accepted: 09/11/2020] [Indexed: 11/22/2022]
Abstract
Ethanolamine is a bioactive molecule found in several cells, including those in the central nervous system (CNS). In the brain, ethanolamine and ethanolamine-related molecules have emerged as prodrug moieties that can promote drug movement across the blood-brain barrier. This improvement in the ability to target drugs to the brain may also mean that in the process ethanolamine concentrations in the brain are increased enough for ethanolamine to exert its own neurological ac-tions. Ethanolamine and its associated products have various positive functions ranging from cell signaling to molecular storage, and alterations in their levels have been linked to neurodegenerative conditions such as Alzheimer's disease. This mini-review focuses on the effects of ethanolamine in the CNS and highlights the possible implications of these effects for drug design.
Collapse
Affiliation(s)
- Asfree Gwanyanya
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town,. South Africa
| | - Christie Nicole Godsmark
- School of Public Health, College of Medicine and Health, University College Cork, Cork,. Ireland
| | - Roisin Kelly-Laubscher
- Department of Pharmacology and Therapeutics, School of Medicine, College of Medicine and Health, University College Cork, Cork,. Ireland
| |
Collapse
|
130
|
Kopfer KH, Jäger W, Matthäus F. A mechanochemical model for rho GTPase mediated cell polarization. J Theor Biol 2020; 504:110386. [DOI: 10.1016/j.jtbi.2020.110386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 01/13/2023]
|
131
|
Rahman K, Coomer CA, Majdoul S, Ding SY, Padilla-Parra S, Compton AA. Homology-guided identification of a conserved motif linking the antiviral functions of IFITM3 to its oligomeric state. eLife 2020; 9:58537. [PMID: 33112230 PMCID: PMC7665892 DOI: 10.7554/elife.58537] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
The interferon-inducible transmembrane (IFITM) proteins belong to the Dispanin/CD225 family and inhibit diverse virus infections. IFITM3 reduces membrane fusion between cells and virions through a poorly characterized mechanism. Mutation of proline-rich transmembrane protein 2 (PRRT2), a regulator of neurotransmitter release, at glycine-305 was previously linked to paroxysmal neurological disorders in humans. Here, we show that glycine-305 and the homologous site in IFITM3, glycine-95, drive protein oligomerization from within a GxxxG motif. Mutation of glycine-95 (and to a lesser extent, glycine-91) disrupted IFITM3 oligomerization and reduced its antiviral activity against Influenza A virus. An oligomerization-defective variant was used to reveal that IFITM3 promotes membrane rigidity in a glycine-95-dependent and amphipathic helix-dependent manner. Furthermore, a compound which counteracts virus inhibition by IFITM3, Amphotericin B, prevented the IFITM3-mediated rigidification of membranes. Overall, these data suggest that IFITM3 oligomers inhibit virus-cell fusion by promoting membrane rigidity.
Collapse
Affiliation(s)
- Kazi Rahman
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, United States
| | - Charles A Coomer
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, United States.,Cellular Imaging Group, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Saliha Majdoul
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, United States
| | - Selena Y Ding
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, United States
| | - Sergi Padilla-Parra
- Cellular Imaging Group, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London, United Kingdom.,Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Alex A Compton
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, United States
| |
Collapse
|
132
|
Hammoud SH, Wehbe Z, Abdelhady S, Kobeissy F, Eid AH, El-Yazbi AF. Dysregulation of Angiotensin Converting Enzyme 2 Expression and Function in Comorbid Disease Conditions Possibly Contributes to Coronavirus Infectious Disease 2019 Complication Severity. Mol Pharmacol 2020; 99:17-28. [PMID: 33082267 DOI: 10.1124/molpharm.120.000119] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/09/2020] [Indexed: 01/08/2023] Open
Abstract
ACE2 has emerged as a double agent in the COVID-19 ordeal, as it is both physiologically protective and virally conducive. The identification of ACE2 in as many as 72 tissues suggests that extrapulmonary invasion and damage is likely, which indeed has already been demonstrated by cardiovascular and gastrointestinal symptoms. On the other hand, identifying ACE2 dysregulation in patients with comorbidities may offer insight as to why COVID-19 symptoms are often more severe in these individuals. This may be attributed to a pre-existing proinflammatory state that is further propelled with the cytokine storm induced by SARS-CoV-2 infection or the loss of functional ACE2 expression as a result of viral internalization. Here, we aim to characterize the distribution and role of ACE2 in various organs to highlight the scope of damage that may arise upon SARS-CoV-2 invasion. Furthermore, by examining the disruption of ACE2 in several comorbid diseases, we offer insight into potential causes of increased severity of COVID-19 symptoms in certain individuals. SIGNIFICANCE STATEMENT: Cell surface expression of ACE2 determines the tissue susceptibility for coronavirus infectious disease 2019 infection. Comorbid disease conditions altering ACE2 expression could increase the patient's vulnerability for the disease and its complications, either directly, through modulation of viral infection, or indirectly, through alteration of inflammatory status.
Collapse
Affiliation(s)
- Safaa H Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Zena Wehbe
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Samar Abdelhady
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Firas Kobeissy
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ali H Eid
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| |
Collapse
|
133
|
Li W, Zhang S, Yang G. Dynamic organization of intracellular organelle networks. WIREs Mech Dis 2020; 13:e1505. [PMID: 32865347 DOI: 10.1002/wsbm.1505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/06/2020] [Accepted: 07/09/2020] [Indexed: 01/07/2023]
Abstract
Intracellular organelles are membrane-bound and biochemically distinct compartments constructed to serve specialized functions in eukaryotic cells. Through extensive interactions, they form networks to coordinate and integrate their specialized functions for cell physiology. A fundamental property of these organelle networks is that they constantly undergo dynamic organization via membrane fusion and fission to remodel their internal connections and to mediate direct material exchange between compartments. The dynamic organization not only enables them to serve critical physiological functions adaptively but also differentiates them from many other biological networks such as gene regulatory networks and cell signaling networks. This review examines this fundamental property of the organelle networks from a systems point of view. The focus is exclusively on homotypic networks formed by mitochondria, lysosomes, endosomes, and the endoplasmic reticulum, respectively. First, key mechanisms that drive the dynamic organization of these networks are summarized. Then, several distinct organizational properties of these networks are highlighted. Next, spatial properties of the dynamic organization of these networks are emphasized, and their functional implications are examined. Finally, some representative molecular machineries that mediate the dynamic organization of these networks are surveyed. Overall, the dynamic organization of intracellular organelle networks is emerging as a fundamental and unifying paradigm in the internal organization of eukaryotic cells. This article is categorized under: Metabolic Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Shuhao Zhang
- Laboratory of Computational Biology and Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, Nankai University, Tianjin, China
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA.,Department of Computational Biology, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
134
|
Dadhich R, Kapoor S. Various Facets of Pathogenic Lipids in Infectious Diseases: Exploring Virulent Lipid-Host Interactome and Their Druggability. J Membr Biol 2020; 253:399-423. [PMID: 32833058 PMCID: PMC7443855 DOI: 10.1007/s00232-020-00135-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
Lipids form an integral, structural, and functional part of all life forms. They play a significant role in various cellular processes such as membrane fusion, fission, endocytosis, protein trafficking, and protein functions. Interestingly, recent studies have revealed their more impactful and critical involvement in infectious diseases, starting with the manipulation of the host membrane to facilitate pathogenic entry. Thereafter, pathogens recruit specific host lipids for the maintenance of favorable intracellular niche to augment their survival and proliferation. In this review, we showcase the lipid-mediated host pathogen interplay in context of life-threatening viral and bacterial diseases including the recent SARS-CoV-2 infection. We evaluate the emergent lipid-centric approaches adopted by these pathogens, while delineating the alterations in the composition and organization of the cell membrane within the host, as well as the pathogen. Lastly, crucial nexus points in their interaction landscape for therapeutic interventions are identified. Lipids act as critical determinants of bacterial and viral pathogenesis by altering the host cell membrane structure and functions.
Collapse
Affiliation(s)
- Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, 400076, India.
- Wadhwani Research Centre for Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| |
Collapse
|
135
|
Akimov SA, Kondrashov OV, Zimmerberg J, Batishchev OV. Ectodomain Pulling Combines with Fusion Peptide Inserting to Provide Cooperative Fusion for Influenza Virus and HIV. Int J Mol Sci 2020; 21:ijms21155411. [PMID: 32751407 PMCID: PMC7432320 DOI: 10.3390/ijms21155411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Enveloped viruses include the most dangerous human and animal pathogens, in particular coronavirus, influenza virus, and human immunodeficiency virus (HIV). For these viruses, receptor binding and entry are accomplished by a single viral envelope protein (termed the fusion protein), the structural changes of which trigger the remodeling and merger of the viral and target cellular membranes. The number of fusion proteins required for fusion activity is still under debate, and several studies report this value to range from 1 to 9 for type I fusion proteins. Here, we consider the earliest stage of viral fusion based on the continuum theory of membrane elasticity. We demonstrate that membrane deformations induced by the oblique insertion of amphipathic fusion peptides mediate the lateral interaction of these peptides and drive them to form into a symmetric fusion rosette. The pulling force produced by the structural rearrangements of the fusion protein ectodomains gives additional torque, which deforms the membrane and additionally stabilizes the symmetric fusion rosette, thus allowing a reduction in the number of fusion peptides needed for fusion. These findings can resolve the large range of published cooperativity indices for HIV, influenza, and other type I fusion proteins.
Collapse
Affiliation(s)
- Sergey A. Akimov
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia; (O.V.K.); (O.V.B.)
- Correspondence: ; Tel.: +7-495-955-4776
| | - Oleg V. Kondrashov
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia; (O.V.K.); (O.V.B.)
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Oleg V. Batishchev
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia; (O.V.K.); (O.V.B.)
| |
Collapse
|
136
|
Tolosa-Díaz A, Almendro-Vedia VG, Natale P, López-Montero I. The GDP-Bound State of Mitochondrial Mfn1 Induces Membrane Adhesion of Apposing Lipid Vesicles through a Cooperative Binding Mechanism. Biomolecules 2020; 10:biom10071085. [PMID: 32708307 PMCID: PMC7407159 DOI: 10.3390/biom10071085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/10/2020] [Accepted: 07/17/2020] [Indexed: 01/01/2023] Open
Abstract
Mitochondria are double-membrane organelles that continuously undergo fission and fusion. Outer mitochondrial membrane fusion is mediated by the membrane proteins mitofusin 1 (Mfn1) and mitofusin 2 (Mfn2), carrying a GTP hydrolyzing domain (GTPase) and two coiled-coil repeats. The detailed mechanism on how the GTP hydrolysis allows Mfns to approach adjacent membranes into proximity and promote their fusion is currently under debate. Using model membranes built up as giant unilamellar vesicles (GUVs), we show here that Mfn1 promotes membrane adhesion of apposing lipid vesicles. The adhesion forces were sustained by the GDP-bound state of Mfn1 after GTP hydrolysis. In contrast, the incubation with the GDP:AlF4−, which mimics the GTP transition state, did not induce membrane adhesion. Due to the flexible nature of lipid membranes, the adhesion strength depended on the surface concentration of Mfn1 through a cooperative binding mechanism. We discuss a possible scenario for the outer mitochondrial membrane fusion based on the modulated action of Mfn1.
Collapse
Affiliation(s)
- Andrés Tolosa-Díaz
- Dto. Química Física, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain; (A.T.-D.); (V.G.A.-V.)
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain
| | - Víctor G. Almendro-Vedia
- Dto. Química Física, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain; (A.T.-D.); (V.G.A.-V.)
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain
| | - Paolo Natale
- Dto. Química Física, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain; (A.T.-D.); (V.G.A.-V.)
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain
- Correspondence: (P.N.); (I.L.-M.)
| | - Iván López-Montero
- Dto. Química Física, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain; (A.T.-D.); (V.G.A.-V.)
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain
- Correspondence: (P.N.); (I.L.-M.)
| |
Collapse
|
137
|
Sora V, Kumar M, Maiani E, Lambrughi M, Tiberti M, Papaleo E. Structure and Dynamics in the ATG8 Family From Experimental to Computational Techniques. Front Cell Dev Biol 2020; 8:420. [PMID: 32587856 PMCID: PMC7297954 DOI: 10.3389/fcell.2020.00420] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/06/2020] [Indexed: 12/31/2022] Open
Abstract
Autophagy is a conserved and essential intracellular mechanism for the removal of damaged components. Since autophagy deregulation is linked to different kinds of pathologies, it is fundamental to gain knowledge on the fine molecular and structural details related to the core proteins of the autophagy machinery. Among these, the family of human ATG8 proteins plays a central role in recruiting other proteins to the different membrane structures involved in the autophagic pathway. Several experimental structures are available for the members of the ATG8 family alone or in complex with their different biological partners, including disordered regions of proteins containing a short linear motif called LC3 interacting motif. Recently, the first structural details of the interaction of ATG8 proteins with biological membranes came into light. The availability of structural data for human ATG8 proteins has been paving the way for studies on their structure-function-dynamic relationship using biomolecular simulations. Experimental and computational structural biology can help to address several outstanding questions on the mechanism of human ATG8 proteins, including their specificity toward different interactors, their association with membranes, the heterogeneity of their conformational ensemble, and their regulation by post-translational modifications. We here summarize the main results collected so far and discuss the future perspectives within the field and the knowledge gaps. Our review can serve as a roadmap for future structural and dynamics studies of the ATG8 family members in health and disease.
Collapse
Affiliation(s)
- Valentina Sora
- Computational Biology Laboratory, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Mukesh Kumar
- Computational Biology Laboratory, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Emiliano Maiani
- Computational Biology Laboratory, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Matteo Lambrughi
- Computational Biology Laboratory, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Matteo Tiberti
- Computational Biology Laboratory, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elena Papaleo
- Computational Biology Laboratory, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
- Translational Disease System Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
138
|
Huang F, Duan R, Zhou Z, Vázquez-González M, Xia F, Willner I. Near-infrared light-activated membrane fusion for cancer cell therapeutic applications. Chem Sci 2020; 11:5592-5600. [PMID: 32874503 PMCID: PMC7441577 DOI: 10.1039/d0sc00863j] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
The spatiotemporal stimulation of liposome-liposome or liposome-membrane fusion processes attracts growing interest as a means to mimic cell-cell interactions in nature and for using these processes for biomedical applications. We report the use of o-nitrobenzyl phosphate functionalized-cholesterol tethered nucleic acid-modified liposomes as functional photoresponsive units for inducing, by NIR-irradiation, spatiotemporal liposome-liposome or liposome-membrane fusion processes. The liposomes are loaded with upconversion nanoparticles (UCNPs) and their NIR irradiation (λ = 980 nm) yields luminescence at λ = 365 nm, providing a localized light-source to deprotect the o-nitrobenzyl phosphate groups and resulting in the fragmentation of the nucleic acid structures. In one system, the NIR-triggered fusion of two liposomes, L1 and L2, is exemplified. Liposome L1 is loaded with UCNPs and Tb3+ ions, and the liposome boundary is functionalized with a cholesterol-tethered, o-nitrobenzyl phosphate caged hairpin nucleic acid structure. Liposome L2 is loaded with 2,6-pyridinedicarboxylic acid, DPA, and its boundary is modified with a cholesterol-tethered nucleic acid, complementary to a part of the caged hairpin, associated with L1. NIR-irradiation of the L1/L2 mixture resulted in the photocleavage of the hairpin structure, associated with L1, and the resulting fragmented nucleic acid associated with L1 hybridized with the nucleic acid linked to L2, leading to the fusion of the two liposomes. The fusion process was followed by dynamic light scattering, and by monitoring the fluorescence of the Tb3+-DPA complex generated upon the fusion of the liposomes and their exchange of contents (fusion efficiency 30%). In a second system, the fusion of the liposomes L1, loaded with UCNPs and doxorubicin (DOX), with HeLa cancer cells functionalized with nucleic acid tethers, complementary to the hairpin units associated with the boundary of L1, and linked to the MUC-1 receptor sites associated with the HeLa cells, through a MUC-1 aptamer unit is exemplified. The effect of DOX-loaded L1/HeLa cell fusion on the cytotoxicity towards HeLa cells is addressed. The NIR UCNP-stimulated cleavage of the o-nitrobenzyl phosphate caged hairpin units associated with L1 leads to the fragmentation of the hairpin units and the resulting nucleic acid tethers hybridize with the nucleic acid-modified HeLa cells, resulting in the liposome-HeLa cell fusion and the release of DOX into the HeLa cells. Selective spatiotemporal cytotoxicity towards HeLa cells is demonstrated (ca. 40% cell killing within two days). The study presents a comprehensive stepwise set of experiments directed towards the development of NIR-driven liposome-liposome or liposome-membrane fusion processes.
Collapse
Affiliation(s)
- Fujian Huang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education , Faculty of Materials Science and Chemistry , China University of Geosciences , Wuhan 430074 , China . ;
| | - Ruilin Duan
- Engineering Research Center of Nano-Geomaterials of Ministry of Education , Faculty of Materials Science and Chemistry , China University of Geosciences , Wuhan 430074 , China . ;
| | - Zhixin Zhou
- Institute of Chemistry , Center for Nanoscience and Nanotechnology , The Hebrew University of Jerusalem , Jerusalem 91904 , Israel .
| | - Margarita Vázquez-González
- Institute of Chemistry , Center for Nanoscience and Nanotechnology , The Hebrew University of Jerusalem , Jerusalem 91904 , Israel .
| | - Fan Xia
- Engineering Research Center of Nano-Geomaterials of Ministry of Education , Faculty of Materials Science and Chemistry , China University of Geosciences , Wuhan 430074 , China . ;
| | - Itamar Willner
- Institute of Chemistry , Center for Nanoscience and Nanotechnology , The Hebrew University of Jerusalem , Jerusalem 91904 , Israel .
| |
Collapse
|
139
|
Tang T, Bidon M, Jaimes JA, Whittaker GR, Daniel S. Coronavirus membrane fusion mechanism offers a potential target for antiviral development. Antiviral Res 2020. [PMID: 32272173 DOI: 10.1016/j.antiviral.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has focused attention on the need to develop effective therapies against the causative agent, SARS-CoV-2, and also against other pathogenic coronaviruses (CoV) that have emerged in the past or might appear in future. Researchers are therefore focusing on steps in the CoV replication cycle that may be vulnerable to inhibition by broad-spectrum or specific antiviral agents. The conserved nature of the fusion domain and mechanism across the CoV family make it a valuable target to elucidate and develop pan-CoV therapeutics. In this article, we review the role of the CoV spike protein in mediating fusion of the viral and host cell membranes, summarizing the results of research on SARS-CoV, MERS-CoV, and recent peer-reviewed studies of SARS-CoV-2, and suggest that the fusion mechanism be investigated as a potential antiviral target. We also provide a supplemental file containing background information on the biology, epidemiology, and clinical features of all human-infecting coronaviruses, along with a phylogenetic tree of these coronaviruses.
Collapse
Affiliation(s)
- Tiffany Tang
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Miya Bidon
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Javier A Jaimes
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, 14853, USA
| | - Gary R Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, 14853, USA
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
140
|
Tang T, Bidon M, Jaimes JA, Whittaker GR, Daniel S. Coronavirus membrane fusion mechanism offers a potential target for antiviral development. Antiviral Res 2020; 178:104792. [PMID: 32272173 PMCID: PMC7194977 DOI: 10.1016/j.antiviral.2020.104792] [Citation(s) in RCA: 521] [Impact Index Per Article: 104.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/05/2020] [Accepted: 04/05/2020] [Indexed: 12/14/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has focused attention on the need to develop effective therapies against the causative agent, SARS-CoV-2, and also against other pathogenic coronaviruses (CoV) that have emerged in the past or might appear in future. Researchers are therefore focusing on steps in the CoV replication cycle that may be vulnerable to inhibition by broad-spectrum or specific antiviral agents. The conserved nature of the fusion domain and mechanism across the CoV family make it a valuable target to elucidate and develop pan-CoV therapeutics. In this article, we review the role of the CoV spike protein in mediating fusion of the viral and host cell membranes, summarizing the results of research on SARS-CoV, MERS-CoV, and recent peer-reviewed studies of SARS-CoV-2, and suggest that the fusion mechanism be investigated as a potential antiviral target. We also provide a supplemental file containing background information on the biology, epidemiology, and clinical features of all human-infecting coronaviruses, along with a phylogenetic tree of these coronaviruses.
Collapse
Affiliation(s)
- Tiffany Tang
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Miya Bidon
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Javier A Jaimes
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, 14853, USA
| | - Gary R Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, 14853, USA
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
141
|
Akimov SA, Molotkovsky RJ, Kuzmin PI, Galimzyanov TR, Batishchev OV. Continuum Models of Membrane Fusion: Evolution of the Theory. Int J Mol Sci 2020; 21:E3875. [PMID: 32485905 PMCID: PMC7312925 DOI: 10.3390/ijms21113875] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/24/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022] Open
Abstract
Starting from fertilization, through tissue growth, hormone secretion, synaptic transmission, and sometimes morbid events of carcinogenesis and viral infections, membrane fusion regulates the whole life of high organisms. Despite that, a lot of fusion processes still lack well-established models and even a list of main actors. A merger of membranes requires their topological rearrangements controlled by elastic properties of a lipid bilayer. That is why continuum models based on theories of membrane elasticity are actively applied for the construction of physical models of membrane fusion. Started from the view on the membrane as a structureless film with postulated geometry of fusion intermediates, they developed along with experimental and computational techniques to a powerful tool for prediction of the whole process with molecular accuracy. In the present review, focusing on fusion processes occurring in eukaryotic cells, we scrutinize the history of these models, their evolution and complication, as well as open questions and remaining theoretical problems. We show that modern approaches in this field allow continuum models of membrane fusion to stand shoulder to shoulder with molecular dynamics simulations, and provide the deepest understanding of this process in multiple biological systems.
Collapse
Affiliation(s)
- Sergey A. Akimov
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia; (R.J.M.); (P.I.K.); (T.R.G.); (O.V.B.)
| | | | | | | | | |
Collapse
|
142
|
Yoshihara A, Watanabe S, Goel I, Ishihara K, Ekdahl KN, Nilsson B, Teramura Y. Promotion of cell membrane fusion by cell-cell attachment through cell surface modification with functional peptide-PEG-lipids. Biomaterials 2020; 253:120113. [PMID: 32438114 DOI: 10.1016/j.biomaterials.2020.120113] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022]
Abstract
Cell fusion is a fundamental event in various biological processes and has been applied to a number of biotechnologies. However, cell fusion efficiency is still low and strongly depends on cell lines and skills, though some improvements have been made. Our hypothesis is that two distinct cell membranes need to be brought together for cell membrane fusion, which is important for mimicking cell fusion in vitro. Here, we aimed to improve the homogeneous and heterogeneous cell fusion efficiency using a cell-cell attachment technique. We modified cellular membranes with two distinctive poly(ethylene glycol)-lipids (PEG-lipids) carrying oligopeptide, three repeated units of the EIAALEK and KIAALKE sequences (fuE3 and fuK3, respectively), which induce cell-cell attachment. The ratio and area of cell-cell attachment can be controlled through surface modification with fuE3-and fuK3-PEG-lipids by changing the number of each incorporated peptide. By combining this technique with the PEG-induced method, the cell fusion efficiency was significantly improved for homogeneous and heterogeneous cell fusion compared to conventional PEG-induced methods. For homogeneous CCRF-CEM cell fusion, the efficiency increased up to 64% from the 8.4% with the PEG-induced method. In addition, for heterogeneous cell fusion of myeloma cells and splenocytes, the efficiency increased up to 18% from almost zero. Thus, cell membrane fusion could be promoted effectively between closely contacted cell membranes induced by the cell-cell attachment technique.
Collapse
Affiliation(s)
- Akifumi Yoshihara
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Sayumi Watanabe
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Isha Goel
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Kazuhiko Ishihara
- Department of Material Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Kristina N Ekdahl
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, SE-391 82, Kalmar, Sweden; Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85, Uppsala, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85, Uppsala, Sweden
| | - Yuji Teramura
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
143
|
Martens S, Fracchiolla D. Activation and targeting of ATG8 protein lipidation. Cell Discov 2020; 6:23. [PMID: 32377373 PMCID: PMC7198486 DOI: 10.1038/s41421-020-0155-1] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
ATG8 family proteins are evolutionary conserved ubiquitin-like modifiers, which become attached to the headgroup of the membrane lipid phosphatidylethanolamine in a process referred to as lipidation. This reaction is carried out analogous to the conjugation of ubiquitin to its target proteins, involving the E1-like ATG7, the E2-like ATG3 and the E3-like ATG12-ATG5-ATG16 complex, which determines the site of lipidation. ATG8 lipidation is a hallmark of autophagy where these proteins are involved in autophagosome formation, the fusion of autophagosomes with lysosomes and cargo selection. However, it has become evident that ATG8 lipidation also occurs in processes that are not directly related to autophagy. Here we discuss recent insights into the targeting of ATG8 lipidation in autophagy and other pathways with special emphasis on the recruitment and activation of the E3-like complex.
Collapse
Affiliation(s)
- Sascha Martens
- Department of Biochemistry and Cell Biology, Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/5, 1030 Vienna, Austria
| | - Dorotea Fracchiolla
- Department of Biochemistry and Cell Biology, Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/5, 1030 Vienna, Austria
| |
Collapse
|
144
|
Fusing Artificial Cell Compartments and Lipid Domains Using Optical Traps: A Tool to Modulate Membrane Composition and Phase Behaviour. MICROMACHINES 2020; 11:mi11040388. [PMID: 32272670 PMCID: PMC7230983 DOI: 10.3390/mi11040388] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/25/2020] [Accepted: 03/28/2020] [Indexed: 01/26/2023]
Abstract
New technologies for manipulating biomembranes have vast potential to aid the understanding of biological phenomena, and as tools to sculpt novel artificial cell architectures for synthetic biology. The manipulation and fusion of vesicles using optical traps is amongst the most promising due to the level of spatiotemporal control it affords. Herein, we conduct a suite of feasibility studies to show the potential of optical trapping technologies to (i) modulate the lipid composition of a vesicle by delivering new membrane material through fusion events and (ii) manipulate and controllably fuse coexisting membrane domains for the first time. We also outline some noteworthy morphologies and transitions that the vesicle undergoes during fusion, which gives us insight into the mechanisms at play. These results will guide future exploitation of laser-assisted membrane manipulation methods and feed into a technology roadmap for this emerging technology.
Collapse
|
145
|
Abstract
Ferlins are multiple-C2-domain proteins involved in Ca2+-triggered membrane dynamics within the secretory, endocytic and lysosomal pathways. In bony vertebrates there are six ferlin genes encoding, in humans, dysferlin, otoferlin, myoferlin, Fer1L5 and 6 and the long noncoding RNA Fer1L4. Mutations in DYSF (dysferlin) can cause a range of muscle diseases with various clinical manifestations collectively known as dysferlinopathies, including limb-girdle muscular dystrophy type 2B (LGMD2B) and Miyoshi myopathy. A mutation in MYOF (myoferlin) was linked to a muscular dystrophy accompanied by cardiomyopathy. Mutations in OTOF (otoferlin) can be the cause of nonsyndromic deafness DFNB9. Dysregulated expression of any human ferlin may be associated with development of cancer. This review provides a detailed description of functions of the vertebrate ferlins with a focus on muscle ferlins and discusses the mechanisms leading to disease development.
Collapse
|
146
|
Branttie JM, Dutch RE. Parainfluenza virus 5 fusion protein maintains pre-fusion stability but not fusogenic activity following mutation of a transmembrane leucine/isoleucine domain. J Gen Virol 2020; 101:467-472. [PMID: 32100701 DOI: 10.1099/jgv.0.001399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The paramyxoviruses Hendra virus (HeV) and parainfluenza virus 5 (PIV5) require the fusion (F) protein to efficiently infect cells. For fusion to occur, F undergoes dramatic, essentially irreversible conformational changes to merge the viral and cell membranes into a continuous bilayer. Recently, a transmembrane (TM) domain leucine/isoleucine (L/I) zipper was shown to be critical in maintaining the expression, stability and pre-fusion conformation of HeV F, allowing for fine-tuned timing of membrane fusion. To analyse the effect of the TM domain L/I zipper in another paramyxovirus, we created alanine mutations to the TM domain of PIV5 F, a paramyxovirus model system. Our data show that while the PIV5 F TM L/I zipper does not significantly affect total expression and only modestly affects surface expression and pre-fusion stability, it is critical for fusogenic activity. These results suggest that the roles of TM L/I zipper motifs differ among members of the family Paramyxoviridae.
Collapse
Affiliation(s)
- Jean Mawuena Branttie
- Department of Molecular and Cellular Biochemistry, College of Medicine University of Kentucky Biomedical Biological Sciences Research Bldg, 741 South Limestone Street, Lexington, KY, USA
| | - Rebecca Ellis Dutch
- Department of Molecular and Cellular Biochemistry, College of Medicine University of Kentucky Biomedical Biological Sciences Research Bldg, 741 South Limestone Street, Lexington, KY, USA
| |
Collapse
|
147
|
French CT, Bulterys PL, Woodward CL, Tatters AO, Ng KR, Miller JF. Virulence from the rhizosphere: ecology and evolution of Burkholderia pseudomallei-complex species. Curr Opin Microbiol 2020; 54:18-32. [PMID: 32028234 DOI: 10.1016/j.mib.2019.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/30/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Christopher T French
- California NanoSystems Institute, UCLA, 570 Westwood Plaza Bldg. 114, 4538 West, Los Angeles, CA 90095, United States; Department of Microbiology, Immunology, and Molecular Genetics, UCLA, 609 Charles E. Young Drive East, Los Angeles, CA 90095, United States; Northern Arizona University, Department of Biological Sciences, Pathogen and Microbiome Institute 1395 S Knoles Drive, Flagstaff, AZ 86011, United States.
| | - Philip L Bulterys
- Department of Pathology, Stanford University, Lane Building, L235, 300 Pasteur Drive, Stanford, CA, 94305, United States
| | - Cora L Woodward
- California NanoSystems Institute, UCLA, 570 Westwood Plaza Bldg. 114, 4538 West, Los Angeles, CA 90095, United States
| | - Avery O Tatters
- California NanoSystems Institute, UCLA, 570 Westwood Plaza Bldg. 114, 4538 West, Los Angeles, CA 90095, United States
| | - Ken R Ng
- California NanoSystems Institute, UCLA, 570 Westwood Plaza Bldg. 114, 4538 West, Los Angeles, CA 90095, United States
| | - Jeff F Miller
- California NanoSystems Institute, UCLA, 570 Westwood Plaza Bldg. 114, 4538 West, Los Angeles, CA 90095, United States; Molecular Biology Institute, UCLA, 611 Charles E. Young Drive East, Los Angeles, CA 90095, United States; Department of Microbiology, Immunology, and Molecular Genetics, UCLA, 609 Charles E. Young Drive East, Los Angeles, CA 90095, United States
| |
Collapse
|
148
|
Sprunck S. Twice the fun, double the trouble: gamete interactions in flowering plants. CURRENT OPINION IN PLANT BIOLOGY 2020; 53:106-116. [PMID: 31841779 DOI: 10.1016/j.pbi.2019.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/14/2019] [Accepted: 11/17/2019] [Indexed: 05/13/2023]
Abstract
During sexual reproduction two gametes of opposite sex unite to produce a zygote. Gamete fusion is a highly controlled process and it has become evident that, across species, common concepts apply to this ancient and fundamental event. Sexual reproduction in flowering plants is even more complex in that two sperm cells fertilize two female reproductive cells (egg and central cell) in a process called double fertilization. Due to the coordinated developmental progression and mutual dependency of the two fertilization products (embryo and endosperm), the success and timing of the two fusion events substantially affects seed set. So far, four proteins are known to act on the surfaces of Arabidopsis gametes to accomplish double fertilization. The molecular and evolutionary characteristics of these players prove that flowering plants integrate plant-specific and widely conserved mechanisms to accomplish the timely fusion of each sperm cell with one female reproductive cell.
Collapse
Affiliation(s)
- Stefanie Sprunck
- Cell Biology and Plant Biochemistry, University of Regensburg, Universitätsstr. 31, 93053 Regensburg, Germany.
| |
Collapse
|
149
|
Zhou J, Shao Z, Liu J, Duan Q, Wang X, Li J, Yang H. From Endocytosis to Nonendocytosis: The Emerging Era of Gene Delivery. ACS APPLIED BIO MATERIALS 2020; 3:2686-2701. [DOI: 10.1021/acsabm.9b01131] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jie Zhou
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Zhentao Shao
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Jia Liu
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Qiao Duan
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Xiang Wang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
| |
Collapse
|
150
|
Yokoyama H, Matsui I. The lipid raft markers stomatin, prohibitin, flotillin, and HflK/C (SPFH)-domain proteins form an operon with NfeD proteins and function with apolar polyisoprenoid lipids. Crit Rev Microbiol 2020; 46:38-48. [PMID: 31983249 DOI: 10.1080/1040841x.2020.1716682] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SPFH-domain proteins are found in almost all organisms across three domains: archaea, bacteria, and eukaryotes. In eukaryotic organelles, their subfamilies exhibit overlapping distribution and functions; thus, the rationality of annotation to discriminate these subfamilies remains unclear. In this review, the binding ability of prokaryotic SPFH-domain proteins towards nonpolar polyisoprenoides such as squalene and lycopene, rather than cholesterol, is discussed. The hydrophobic region at the C-terminus of SPFH-domain proteins constitutes the main region that binds apolar polyisoprenoid lipids as well as cholesterol and substantively contributes towards lipid raft formation as these regions are self-assembled together with specific lipids. Because the scaffolding proteins caveolins show common topological properties with SPFH-domain proteins such as stomatin and flotillin, the α-helical segments of stomatin proteins can flexibly move along with the membrane surface, with such movement potentially leading to membrane bending via lipid raft clustering through the formation of high order homo-oligomeric complexes of SPFH-domain proteins. We also discuss the functional significance and ancient origin of SPFH-domain proteins and the NfeD protein (STOPP) operon, which can be traced back to the ancient living cells that diverged and evolved to archaea and bacteria. Based on the molecular mechanism whereby the STOPP-protease degrades the C-terminal hydrophobic clusters of SPFH-domain proteins, it is conceivable that STOPP-protease might control the physicochemical properties of lipid rafts.
Collapse
Affiliation(s)
- Hideshi Yokoyama
- Department of Medical and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Ikuo Matsui
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|