51
|
Miyata Y, Yamada K, Nagata S, Segawa K. Two types of type IV P-type ATPases independently re-establish the asymmetrical distribution of phosphatidylserine in plasma membranes. J Biol Chem 2022; 298:102527. [PMID: 36162506 PMCID: PMC9597894 DOI: 10.1016/j.jbc.2022.102527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
Phospholipids are asymmetrically distributed between the lipid bilayer of plasma membranes in which phosphatidylserine (PtdSer) is confined to the inner leaflet. ATP11A and ATP11C, type IV P-Type ATPases in plasma membranes, flip PtdSer from the outer to the inner leaflet, but involvement of other P4-ATPases is unclear. We herein demonstrated that once PtdSer was exposed on the cell surface of ATP11A−/−ATP11C−/− mouse T cell line (W3), its internalization to the inner leaflet of plasma membranes was negligible at 15 °C. However, ATP11A−/−ATP11C−/− cells internalized the exposed PtdSer at 37 °C, a temperature at which trafficking of intracellular membranes was active. In addition to ATP11A and 11C, W3 cells expressed ATP8A1, 8B2, 8B4, 9A, 9B, and 11B, with ATP8A1 and ATP11B being present at recycling endosomes. Cells deficient in four P4-ATPases (ATP8A1, 11A, 11B, and 11C) (QKO) did not constitutively expose PtdSer on the cell surface but lost the ability to re-establish PtdSer asymmetry within 1 hour, even at 37 °C. The expression of ATP11A or ATP11C conferred QKO cells with the ability to rapidly re-establish PtdSer asymmetry at 15 °C and 37 °C, while cells expressing ATP8A1 or ATP11B required a temperature of 37 °C to achieve this function, and a dynamin inhibitor blocked this process. These results revealed that mammalian cells are equipped with two independent mechanisms to re-establish its asymmetry: the first is a rapid process involving plasma membrane flippases, ATP11A and ATP11C, while the other is mediated by ATP8A1 and ATP11B, which require an endocytosis process.
Collapse
Affiliation(s)
- Yugo Miyata
- Department of Medical Chemistry, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Kyoko Yamada
- Laboratory of Biochemistry & Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Shigekazu Nagata
- Laboratory of Biochemistry & Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
| | - Katsumori Segawa
- Department of Medical Chemistry, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan; Laboratory of Biochemistry & Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
52
|
The function of Piezo1 in hepatoblastoma metastasis and its potential transduction mechanism. Heliyon 2022; 8:e10301. [PMID: 36097495 PMCID: PMC9463386 DOI: 10.1016/j.heliyon.2022.e10301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/25/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Background Hepatoblastoma (HB) is the most common primary malignant liver tumor in children. The prognosis of HB metastasis is poor, despite the increasing diversity of treatment. Piezo, a ubiquitously expressed membrane mechano-transduction protein, is involved in the process of tumor cell migration. Under the gene expression profiling interactive analysis (GEPIA) database, Piezo1 was highly expressed in HB and negatively correlated with the overall survival time. Methods Firstly, the expression of Piezo1 in both paracancerous and HB tissues (n = 7) was detected, and the prognostic value of Piezo1 was assessed in HB (n = 160) patients. Secondly, the inhibition and overexpression of Piezo1were executed in two HB cell lines, HepG2 and Huh 6. Methyl thiazolyl tetrazolium (MTT), wound healing and trans-well assays were performed to identify the effect of Piezo1 on the proliferation and metastasis of HB cells, respectively. In addition, a co-immunoprecipitation assay was performed to determine whether Piezo1 has an interaction with HIF-1α. Finally, the expressions level of Piezo1, HIF-1α, and VEGF by overexpression/inhibition each other were detected by RT-qPCR and western blots to find a possible signaling channel in HB metastasis. Results We found that Piezo1 was highly expressed in HB tissues and associated with poor prognosis of patients. Piezo1 was related to cell proliferation in HepG2 and Huh 6 cells. We also found that Piezo1 stimulated HIF-1α expression. Meanwhile, overexpression of Piezo1 promoted the migration and invasion of HB cells, while the promotion was not detected when HIF-1α was suppressed. Additionally, the silencing of HIF-1α inhibited the expression of VEGF, but showed no effect on Piezo1 expression. Conclusion In this study, we identified that Piezo1 was involved in HB metastasis, and the Piezo1-HIF-1α-VEGF axis could be a possible signaling pathway in HB metastasis.
Collapse
|
53
|
Huang J, Zhang K, Du R, Liu W, Zhang H, Tian T, Wang Y, Wang G, Yin T. The Janus-faced role of Piezo1 in cardiovascular health under mechanical stimulation. Genes Dis 2022. [PMID: 37492728 PMCID: PMC10363580 DOI: 10.1016/j.gendis.2022.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
In recent years, cardiovascular health problems are becoming more and more serious. At the same time, mechanical stimulation closely relates to cardiovascular health. In this context, Piezo1, which is very sensitive to mechanical stimulation, has attracted our attention. Here, we review the critical significance of Piezo1 in mechanical stimulation of endothelial cells, NO production, lipid metabolism, DNA damage protection, the development of new blood vessels and maturation, narrowing of blood vessels, blood pressure regulation, vascular permeability, insulin sensitivity, and maintenance of red blood cell function. Besides, Piezo1 may participate in the occurrence and development of atherosclerosis, diabetes, hypertension, and other cardiovascular diseases. It is worth noting that Piezo1 has dual effects on maintaining cardiovascular health. On the one hand, the function of Piezo1 is necessary to maintain cardiovascular health; on the other hand, under some extreme mechanical stimulation, the overexpression of Piezo1 may bring adverse factors such as inflammation. Therefore, this review discusses the Janus-faced role of Piezo1 in maintaining cardiovascular health and puts forward new ideas to provide references for gene therapy or nanoagents targeting Piezo1.
Collapse
|
54
|
Lobba ARM, Alvarez-Flores MP, Fessel MR, Buri MV, Oliveira DS, Gomes RN, Cunegundes PS, DeOcesano-Pereira C, Cinel VD, Chudzinski-Tavassi AM. A Kunitz-type inhibitor from tick salivary glands: A promising novel antitumor drug candidate. Front Mol Biosci 2022; 9:936107. [PMID: 36052162 PMCID: PMC9424826 DOI: 10.3389/fmolb.2022.936107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Salivary glands are vital structures responsible for successful tick feeding. The saliva of ticks contains numerous active molecules that participate in several physiological processes. A Kunitz-type factor Xa (FXa) inhibitor, similar to the tissue factor pathway inhibitor (TFPI) precursor, was identified in the salivary gland transcriptome of Amblyomma sculptum ticks. The recombinant mature form of this Kunitz-type inhibitor, named Amblyomin-X, displayed anticoagulant, antiangiogenic, and antitumor properties. Amblyomin-X is a protein that inhibits FXa in the blood coagulation cascade and acts via non-hemostatic mechanisms, such as proteasome inhibition. Amblyomin-X selectively induces apoptosis in cancer cells and promotes tumor regression through these mechanisms. Notably, the cytotoxicity of Amblyomin-X seems to be restricted to tumor cells and does not affect non-tumorigenic cells, tissues, and organs, making this recombinant protein an attractive molecule for anticancer therapy. The cytotoxic activity of Amblyomin-X on tumor cells has led to vast exploration into this protein. Here, we summarize the function, action mechanisms, structural features, pharmacokinetics, and biodistribution of this tick Kunitz-type inhibitor recombinant protein as a promising novel antitumor drug candidate.
Collapse
Affiliation(s)
- Aline R. M. Lobba
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Miryam Paola Alvarez-Flores
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Melissa Regina Fessel
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Marcus Vinicius Buri
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Douglas S. Oliveira
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
- Biochemistry Department, Federal University of São Paulo, São Paulo, Brazil
| | - Renata N. Gomes
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Priscila S. Cunegundes
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
- Biochemistry Department, Federal University of São Paulo, São Paulo, Brazil
| | - Carlos DeOcesano-Pereira
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
| | - Victor D. Cinel
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
- Biochemistry Department, Federal University of São Paulo, São Paulo, Brazil
| | - Ana M. Chudzinski-Tavassi
- Centre of Excellence in New Target Discovery-CENTD, Butantan Institute, São Paulo, Brazil
- Development and Innovation Centre, Butantan Institute, Butantan Institute, São Paulo, Brazil
- Biochemistry Department, Federal University of São Paulo, São Paulo, Brazil
- *Correspondence: Ana M. Chudzinski-Tavassi,
| |
Collapse
|
55
|
Buyan A, Allender DW, Corry B, Schick M. Lipid redistribution in the highly curved footprint of Piezo1. Biophys J 2022:S0006-3495(22)00595-1. [PMID: 35927961 DOI: 10.1016/j.bpj.2022.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/06/2022] [Accepted: 07/18/2022] [Indexed: 11/15/2022] Open
Abstract
We investigate the effects on the distribution of lipids in the plasma membrane that are caused by the insertion of a protein, Piezo1, that significantly distorts the membrane toward the cytosol. From coarse-grained molecular dynamics simulations, we find that the major effects occur in the outer, extracellular, leaflet. The mol fraction of cholesterol increases significantly in the curved region of the membrane close to Piezo1, while those of phosphatidylcholine and of sphingomyelin decrease. In the inner leaflet, mol fractions of cholesterol and of phosphatidylethanolamine decrease slightly as the protein is approached, while that of phosphatidylserine increases slightly. The mol fraction of phosphatidylcholine decreases markedly as the protein is approached. Most of these results are understood in the context of a theoretical model that utilizes two elements; (i) a coupling between the leaflets' actual curvatures and their compositionally-dependent spontaneous curvatures and, (ii) the dependence of the spontaneous curvatures not only on the mol fractions of the phospholipids, but also on the effect that cholesterol has on the spontaneous curvatures of the phospholipids.
Collapse
Affiliation(s)
- Amanda Buyan
- Research School of Biology, The Australian National University, Acton, Australia
| | - D W Allender
- Department of Physics, University of Washington, Seattle WA; Department of Physics, Kent State University, Kent OH
| | - Ben Corry
- Research School of Biology, The Australian National University, Acton, Australia
| | - M Schick
- Department of Physics, University of Washington, Seattle WA
| |
Collapse
|
56
|
Szondy Z, Al‐Zaeed N, Tarban N, Fige É, Garabuczi É, Sarang Z. Involvement of phosphatidylserine receptors in the skeletal muscle regeneration: therapeutic implications. J Cachexia Sarcopenia Muscle 2022; 13:1961-1973. [PMID: 35666022 PMCID: PMC9397555 DOI: 10.1002/jcsm.13024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 04/09/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Sarcopenia is a progressive loss of muscle mass and strength with a risk of adverse outcomes such as disability, poor quality of life, and death. Increasing evidence indicates that diminished ability of the muscle to activate satellite cell-dependent regeneration is one of the factors that might contribute to its development. Skeletal muscle regeneration following myogenic cell death results from the proliferation and differentiation of myogenic stem cells, called satellite cells, located beneath the basal lamina of the muscle fibres. Satellite cell differentiation is not a satellite cell-autonomous process but depends on signals provided by the surrounding cells. Infiltrating macrophages play a key role in the process partly by clearing the necrotic cell debris, partly by producing cytokines and growth factors that guide myogenesis. At the beginning of the muscle regeneration process, macrophages are pro-inflammatory, and the cytokines produced by them trigger the proliferation and differentiation of satellite cells. Following the uptake of dead cells, however, a transcriptionally regulated phenotypic change (macrophage polarization) is induced in them resulting in their transformation into healing macrophages that guide resolution of inflammation, completion of myoblast differentiation, myoblast fusion and growth, and return to homeostasis. Impaired efferocytosis results in delayed cell death clearance, delayed macrophage polarization, prolonged inflammation, and impaired muscle regeneration. Thus, proper efferocytosis by macrophages is a determining factor during muscle repair. Here we review that both efferocytosis and myogenesis are dependent on the cell surface phosphatidylserine (PS), and surprisingly, these two processes share a number of common PS receptors and signalling pathways. Based on these findings, we propose that stimulating the function of PS receptors for facilitating muscle repair following injury could be a successful approach, as it would enhance efferocytosis and myogenesis simultaneously. Because increasing evidence indicates a pathophysiological role of impaired efferocytosis in the development of chronic inflammatory conditions, as well as in impaired muscle regeneration both contributing to the development of sarcopenia, improving efferocytosis should be considered also in its management. Again applying or combining those treatments that target PS receptors would be expected to be the most effective, because they would also promote myogenesis. A potential PS receptor-triggering candidate molecule is milk fat globule-EGF-factor 8 (MFG-E8), which not only stimulates PS-dependent efferocytosis and myoblast fusion but also promotes extracellular signal-regulated kinase (ERK) and Akt activation-mediated cell proliferation and cell cycle progression in myoblasts.
Collapse
Affiliation(s)
- Zsuzsa Szondy
- Section of Dental Biochemistry, Department of Basic Medical Sciences, Faculty of DentistryUniversity of DebrecenDebrecenHungary
- Department of Biochemistry and Molecular Biology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Nour Al‐Zaeed
- Doctoral School of Molecular Cell and Immune Biology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Nastaran Tarban
- Doctoral School of Molecular Cell and Immune Biology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Éva Fige
- Section of Dental Biochemistry, Department of Basic Medical Sciences, Faculty of DentistryUniversity of DebrecenDebrecenHungary
| | - Éva Garabuczi
- Department of Biochemistry and Molecular Biology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| |
Collapse
|
57
|
Young M, Lewis AH, Grandl J. Physics of mechanotransduction by Piezo ion channels. J Gen Physiol 2022; 154:213231. [PMID: 35593732 PMCID: PMC9127981 DOI: 10.1085/jgp.202113044] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/28/2022] [Accepted: 04/29/2022] [Indexed: 12/26/2022] Open
Abstract
Piezo ion channels are sensors of mechanical forces and mediate a wide range of physiological mechanotransduction processes. More than a decade of intense research has elucidated much of the structural and mechanistic principles underlying Piezo gating and its roles in physiology, although wide gaps of knowledge continue to exist. Here, we review the forces and energies involved in mechanical activation of Piezo ion channels and their functional modulation by other chemical and physical stimuli including lipids, voltage, and temperature. We compare the three predominant mechanisms likely to explain Piezo activation—the force-from-lipids mechanism, the tether model, and the membrane footprint theory. Additional sections shine light on how Piezo ion channels may affect each other through spatial clustering and functional cooperativity, and how substantial functional heterogeneity of Piezo ion channels arises as a byproduct of the precise physical environment each channel experiences. Finally, our review concludes by pointing out major research questions and technological limitations that future research can address.
Collapse
Affiliation(s)
- Michael Young
- Department of Neurobiology, Duke University Medical Center, Durham, NC
| | - Amanda H Lewis
- Department of Neurobiology, Duke University Medical Center, Durham, NC
| | - Jörg Grandl
- Department of Neurobiology, Duke University Medical Center, Durham, NC
| |
Collapse
|
58
|
Millay DP. Regulation of the myoblast fusion reaction for muscle development, regeneration, and adaptations. Exp Cell Res 2022; 415:113134. [PMID: 35367215 PMCID: PMC9058940 DOI: 10.1016/j.yexcr.2022.113134] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/23/2022] [Accepted: 03/28/2022] [Indexed: 12/27/2022]
Abstract
Fusion of plasma membranes is essential for skeletal muscle development, regeneration, exercise-induced adaptations, and results in a cell that contains hundreds to thousands of nuclei within a shared cytoplasm. The differentiation process in myocytes culminates in their fusion to form a new myofiber or fusion to an existing myofiber thereby contributing more synthetic material to the syncytium. The choice for two cells to fuse and become one could be a dangerous event if the two cells are not committed to an allied function. Thus, fusion events are highly regulated with positive and negative factors to fine-tune the process, and requires muscle-specific fusogens (Myomaker and Myomerger) as well as general cellular machinery to achieve the union of membranes. While a unified vertebrate myoblast fusion pathway is not yet established, recent discoveries should make this pursuit attainable. Not only does myocyte fusion impact the normal biology of skeletal muscle, but new evidence indicates dysregulation of the process impacts pathologies of skeletal muscle. Here, I will highlight the molecular players and biochemical mechanisms that drive fusion events in muscle, and discuss how this key myogenic process impacts skeletal muscle diseases.
Collapse
Affiliation(s)
- Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| |
Collapse
|
59
|
Bernareggi A, Bosutti A, Massaria G, Giniatullin R, Malm T, Sciancalepore M, Lorenzon P. The State of the Art of Piezo1 Channels in Skeletal Muscle Regeneration. Int J Mol Sci 2022; 23:ijms23126616. [PMID: 35743058 PMCID: PMC9224226 DOI: 10.3390/ijms23126616] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 02/07/2023] Open
Abstract
Piezo1 channels are highly mechanically-activated cation channels that can sense and transduce the mechanical stimuli into physiological signals in different tissues including skeletal muscle. In this focused review, we summarize the emerging evidence of Piezo1 channel-mediated effects in the physiology of skeletal muscle, with a particular focus on the role of Piezo1 in controlling myogenic precursor activity and skeletal muscle regeneration and vascularization. The disclosed effects reported by pharmacological activation of Piezo1 channels with the selective agonist Yoda1 indicate a potential impact of Piezo1 channel activity in skeletal muscle regeneration, which is disrupted in various muscular pathological states. All findings reported so far agree with the idea that Piezo1 channels represent a novel, powerful molecular target to develop new therapeutic strategies for preventing or ameliorating skeletal muscle disorders characterized by an impairment of tissue regenerative potential.
Collapse
Affiliation(s)
- Annalisa Bernareggi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
- Correspondence:
| | - Alessandra Bosutti
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
| | - Gabriele Massaria
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (R.G.); (T.M.)
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (R.G.); (T.M.)
| | - Marina Sciancalepore
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
| | - Paola Lorenzon
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
| |
Collapse
|
60
|
Benedetto A, Kelley EG. Absorption of the [bmim][Cl] Ionic Liquid in DMPC Lipid Bilayers across Their Gel, Ripple, and Fluid Phases. J Phys Chem B 2022; 126:3309-3318. [PMID: 35472281 PMCID: PMC9082605 DOI: 10.1021/acs.jpcb.2c00710] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/11/2022] [Indexed: 12/19/2022]
Abstract
Lipid bilayers are a key component of cell membranes and play a crucial role in life and in bio-nanotechnology. As a result, controlling their physicochemical properties holds the promise of effective therapeutic strategies. Ionic liquids (ILs)─a vast class of complex organic electrolytes─have shown a high degree of affinity with lipid bilayers and can be exploited in this context. However, the chemical physics of IL absorption and partitioning into lipid bilayers is yet to be fully understood. This work focuses on the absorption of the model IL [bmim][Cl] into 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) lipid bilayers across their gel, ripple, and fluid phases. Here, by small-angle neutron scattering, we show that (i) the IL cations are absorbed in the lipid bilayer in all its thermodynamic phases and (ii) the amount of IL inserted into the lipid phase increased with increasing temperature, changing from three to four IL cations per 10 lipids with increasing temperature from 10 °C in the gel phase to 40 °C in the liquid phase, respectively. An explicative hypothesis, based on the entropy gain coming from the IL hydration water, is presented to explain the observed temperature trend. The ability to control IL absorption with temperature can be used as a handle to tune the effect of ILs on biomembranes and can be exploited in bio-nanotechnological applications.
Collapse
Affiliation(s)
- Antonio Benedetto
- Department
of Science, University of Roma Tre, 00146 Rome, Italy
- School
of Physics, and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
- Laboratory
for Neutron Scattering, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Elizabeth G. Kelley
- NIST
Center for Neutron Research, National Institute
of Standards and Technology, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
61
|
Inefficient development of syncytiotrophoblasts in the Atp11a-deficient mouse placenta. Proc Natl Acad Sci U S A 2022; 119:e2200582119. [PMID: 35476530 PMCID: PMC9170144 DOI: 10.1073/pnas.2200582119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Plasma membranes are composed of a lipid bilayer in which phosphatidylserine (PtdSer) is confined to the inner leaflet by the action of flippase that translocates PtdSer from the outer to inner leaflets. Two P4-ATPases (ATP11A and ATP11C) work as flippase at plasma membranes. Here, we report that the mouse placenta expresses only ATP11A, and Atp11a-deficient mouse embryos die during embryogenesis due to inefficient formation of syncytiotrophoblasts in the placental labyrinth. The flippase-null mutation inactivates human choriocarcinoma BeWo cells to translocate PtdSer into the inner leaflet and undergo cell fusion. These findings highlight the importance of flippase to regulate the distribution of phospholipids for cell fusion, at least in trophoblast fusion. The P4-ATPases ATP11A and ATP11C function as flippases at the plasma membrane to translocate phosphatidylserine from the outer to the inner leaflet. We herein demonstrated that Atp11a-deficient mouse embryos died at approximately E14.5 with thin-walled heart ventricles. However, the cardiomyocyte- or epiblast-specific Atp11a deletion did not affect mouse development or mortality. ATP11C may have compensated for the function of ATP11A in most of the cell types in the embryo. On the other hand, Atp11a, but not Atp11c, was expressed in the mouse placenta, and the Atp11a-null mutation caused poor development of the labyrinthine layer with an increased number of TUNEL-positive foci. Immunohistochemistry and electron microscopy revealed a disorganized labyrinthine layer with unfused trophoblasts in the Atp11a-null placenta. Human placenta-derived choriocarcinoma BeWo cells expressed the ATP11A and ATP11C genes. A lack of ATP11A and ATP11C eliminated the ability of BeWo cells to flip phosphatidylserine and fuse when treated with forskolin. These results indicate that flippases at the plasma membrane play an important role in the formation of syncytiotrophoblasts in placental development.
Collapse
|
62
|
Miles L, Powell J, Kozak C, Song Y. Mechanosensitive Ion Channels, Axonal Growth, and Regeneration. Neuroscientist 2022:10738584221088575. [PMID: 35414308 PMCID: PMC9556659 DOI: 10.1177/10738584221088575] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cells sense and respond to mechanical stimuli by converting those stimuli into biological signals, a process known as mechanotransduction. Mechanotransduction is essential in diverse cellular functions, including tissue development, touch sensitivity, pain, and neuronal pathfinding. In the search for key players of mechanotransduction, several families of ion channels were identified as being mechanosensitive and were demonstrated to be activated directly by mechanical forces in both the membrane bilayer and the cytoskeleton. More recently, Piezo ion channels were discovered as a bona fide mechanosensitive ion channel, and its characterization led to a cascade of research that revealed the diverse functions of Piezo proteins and, in particular, their involvement in neuronal repair.
Collapse
Affiliation(s)
- Leann Miles
- The Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jackson Powell
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Casey Kozak
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuanquan Song
- The Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania, Philadelphia, PA, USA.,Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
63
|
Peng Y, Du J, Günther S, Guo X, Wang S, Schneider A, Zhu L, Braun T. Mechano-signaling via Piezo1 prevents activation and p53-mediated senescence of muscle stem cells. Redox Biol 2022; 52:102309. [PMID: 35395625 PMCID: PMC9005960 DOI: 10.1016/j.redox.2022.102309] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/20/2022] [Accepted: 03/30/2022] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle stem cells (MuSCs), also called satellite cells, are instrumental for postnatal muscle growth and skeletal muscle regeneration. Numerous signaling cascades regulate the fate of MuSCs during muscle regeneration but the molecular mechanism by which MuSCs sense mechanical stimuli remain unclear. Here, we describe that Piezo1, a mechanosensitive ion channel, keeps MuSCs in a quiescent state and prevents senescence. Absence of Piezo1 induces precocious activation of MuSCs, attenuates proliferation, and impairs differentiation, essentially abolishing efficient skeletal muscle regeneration and replenishment of the MuSC pool. Furthermore, we discovered that inactivation of Piezo1 results in compensatory up-regulation of T-type voltage-gated Ca2+ channels, leading to increased Ca2+ influx, which strongly induces NOX4 expression via cPKC. Elevated NOX4 expression in Piezo1-deficient MuSCs increases ROS levels and DNA damage, causing P53-dependent cellular senescence and cell death. The importance of the P53/P21-axis for mediating Piezo1-dependent cellular defects was confirmed by pharmacological inhibition of P53 in Piezo1-deficient mice, which abrogates increased senescence of muscle cells and normalizes muscle regeneration. Our findings uncover an essential role of Piezo1-mediated mechano-signaling in MuSCs for maintaining quiescence and preventing senescence. Reduced mechano-signaling due to decreased physical activity during aging may contribute to the increase of senescent cells and the decline of MuSC numbers in geriatric mice and humans. Piezo1 is highly expressed in skeletal MuSCs and prevents their precocious activation. Loss of Piezo1 increases Ca2+ influx into MuSCs, which induces NOX4 expression via PKC, leading to enhanced ROS generation. Inactivation of Piezo1 depletes the MuSC pool and causes P53-dependent senescence of MuSCs. ROS scavenging in Piezo1-deficient MuSCs prevents P53 accumulation. Inhibition of P53 mitigates skeletal muscle regeneration defects in mice with Piezo1-deficient MuSCs.
Collapse
Affiliation(s)
- Yundong Peng
- Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, 61231, Bad Nauheim, Germany
| | - Jingjing Du
- Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, 61231, Bad Nauheim, Germany; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Stefan Günther
- Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, 61231, Bad Nauheim, Germany
| | - Xinyue Guo
- Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, 61231, Bad Nauheim, Germany
| | - Shengpeng Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, No.76 West Yanta Road, Yanta District, Xi'an, China
| | - Andre Schneider
- Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, 61231, Bad Nauheim, Germany
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Thomas Braun
- Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, 61231, Bad Nauheim, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt am Main, Germany.
| |
Collapse
|
64
|
Ma N, Chen D, Lee JH, Kuri P, Hernandez EB, Kocan J, Mahmood H, Tichy ED, Rompolas P, Mourkioti F. Piezo1 regulates the regenerative capacity of skeletal muscles via orchestration of stem cell morphological states. SCIENCE ADVANCES 2022; 8:eabn0485. [PMID: 35302846 PMCID: PMC8932657 DOI: 10.1126/sciadv.abn0485] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/26/2022] [Indexed: 05/08/2023]
Abstract
Muscle stem cells (MuSCs) are essential for tissue homeostasis and regeneration, but the potential contribution of MuSC morphology to in vivo function remains unknown. Here, we demonstrate that quiescent MuSCs are morphologically heterogeneous and exhibit different patterns of cellular protrusions. We classified quiescent MuSCs into three functionally distinct stem cell states: responsive, intermediate, and sensory. We demonstrate that the shift between different stem cell states promotes regeneration and is regulated by the sensing protein Piezo1. Pharmacological activation of Piezo1 is sufficient to prime MuSCs toward more responsive cells. Piezo1 deletion in MuSCs shifts the distribution toward less responsive cells, mimicking the disease phenotype we find in dystrophic muscles. We further demonstrate that Piezo1 reactivation ameliorates the MuSC morphological and regenerative defects of dystrophic muscles. These findings advance our fundamental understanding of how stem cells respond to injury and identify Piezo1 as a key regulator for adjusting stem cell states essential for regeneration.
Collapse
Affiliation(s)
- Nuoying Ma
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bioengineering Graduate Program, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Delia Chen
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ji-Hyung Lee
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paola Kuri
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward Blake Hernandez
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jacob Kocan
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamd Mahmood
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elisia D. Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Panteleimon Rompolas
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
65
|
Ganassi M, Zammit PS. Involvement of muscle satellite cell dysfunction in neuromuscular disorders: Expanding the portfolio of satellite cell-opathies. Eur J Transl Myol 2022; 32:10064. [PMID: 35302338 PMCID: PMC8992676 DOI: 10.4081/ejtm.2022.10064] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/11/2022] [Indexed: 12/03/2022] Open
Abstract
Neuromuscular disorders are a heterogeneous group of acquired or hereditary conditions that affect striated muscle function. The resulting decrease in muscle strength and motility irreversibly impacts quality of life. In addition to directly affecting skeletal muscle, pathogenesis can also arise from dysfunctional crosstalk between nerves and muscles, and may include cardiac impairment. Muscular weakness is often progressive and paralleled by continuous decline in the ability of skeletal muscle to functionally adapt and regenerate. Normally, the skeletal muscle resident stem cells, named satellite cells, ensure tissue homeostasis by providing myoblasts for growth, maintenance, repair and regeneration. We recently defined 'Satellite Cell-opathies' as those inherited neuromuscular conditions presenting satellite cell dysfunction in muscular dystrophies and myopathies (doi:10.1016/j.yexcr.2021.112906). Here, we expand the portfolio of Satellite Cell-opathies by evaluating the potential impairment of satellite cell function across all 16 categories of neuromuscular disorders, including those with mainly neurogenic and cardiac involvement. We explore the expression dynamics of myopathogenes, genes whose mutation leads to skeletal muscle pathogenesis, using transcriptomic analysis. This revealed that 45% of myopathogenes are differentially expressed during early satellite cell activation (0 - 5 hours). Of these 271 myopathogenes, 83 respond to Pax7, a master regulator of satellite cells. Our analysis suggests possible perturbation of satellite cell function in many neuromuscular disorders across all categories, including those where skeletal muscle pathology is not predominant. This characterisation further aids understanding of pathomechanisms and informs on development of prognostic and diagnostic tools, and ultimately, new therapeutics.
Collapse
Affiliation(s)
- Massimo Ganassi
- King's College London, Randall Centre for Cell and Molecular Biophysics, Guy's Campus, London.
| | - Peter S Zammit
- King's College London, Randall Centre for Cell and Molecular Biophysics, Guy's Campus, London.
| |
Collapse
|
66
|
Savitskaya MA, Zakharov II, Onishchenko GE. Apoptotic Features in Non-Apoptotic Processes. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:191-206. [PMID: 35526851 DOI: 10.1134/s0006297922030014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 01/02/2022] [Accepted: 01/15/2022] [Indexed: 06/14/2023]
Abstract
Apoptosis is the most thoroughly studied type of regulated cell death. Certain events, such as externalization of phosphatidylserine (PS) into the outer leaflet of plasma membrane, mitochondrial outer membrane permeabilization, caspase cascade activation, DNA fragmentation and blebbing, are widely considered to be hallmarks of apoptosis as well as being traditionally viewed as irreversible. This review shows that under particular circumstances these events can also participate in physiological processes not associated with initiation of apoptosis, such as cell differentiation, division, and motility, as well as non-apoptotic types of cell death. Moreover, these events may often be reversible. This review focuses on three processes: phosphatidylserine externalization, blebbing, and activation of apoptotic caspases. Mitochondrial outer membrane permeabilization and DNA fragmentation are not discussed.
Collapse
Affiliation(s)
| | - Ilya I Zakharov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | | |
Collapse
|
67
|
Bartoli F, Debant M, Chuntharpursat-Bon E, Evans EL, Musialowski KE, Parsonage G, Morley LC, Futers TS, Sukumar P, Bowen TS, Kearney MT, Lichtenstein L, Roberts LD, Beech DJ. Endothelial Piezo1 sustains muscle capillary density and contributes to physical activity. J Clin Invest 2022; 132:141775. [PMID: 35025768 PMCID: PMC8884896 DOI: 10.1172/jci141775] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/11/2022] [Indexed: 11/17/2022] Open
Abstract
Piezo1 forms mechanically activated nonselective cation channels that contribute to endothelial response to fluid flow. Here we reveal an important role in the control of capillary density. Conditional endothelial cell-specific deletion of Piezo1 in adult mice depressed physical performance. Muscle microvascular endothelial cell apoptosis and capillary rarefaction were evident and sufficient to account for the effect on performance. There was selective upregulation of thrombospondin-2 (TSP2), an inducer of endothelial cell apoptosis, with no effect on TSP1, a related important player in muscle physiology. TSP2 was poorly expressed in muscle endothelial cells but robustly expressed in muscle pericytes, in which nitric oxide (NO) repressed the Tsp2 gene without an effect on Tsp1. In endothelial cells, Piezo1 was required for normal expression of endothelial NO synthase. The data suggest an endothelial cell-pericyte partnership of muscle in which endothelial Piezo1 senses blood flow to sustain capillary density and thereby maintain physical capability.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - T. Scott Bowen
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | | | | | | | | |
Collapse
|
68
|
Attwaters M, Hughes SM. Cellular and molecular pathways controlling muscle size in response to exercise. FEBS J 2022; 289:1428-1456. [PMID: 33755332 DOI: 10.1111/febs.15820] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/27/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022]
Abstract
From the discovery of ATP and motor proteins to synaptic neurotransmitters and growth factor control of cell differentiation, skeletal muscle has provided an extreme model system in which to understand aspects of tissue function. Muscle is one of the few tissues that can undergo both increase and decrease in size during everyday life. Muscle size depends on its contractile activity, but the precise cellular and molecular pathway(s) by which the activity stimulus influences muscle size and strength remain unclear. Four correlates of muscle contraction could, in theory, regulate muscle growth: nerve-derived signals, cytoplasmic calcium dynamics, the rate of ATP consumption and physical force. Here, we summarise the evidence for and against each stimulus and what is known or remains unclear concerning their molecular signal transduction pathways and cellular effects. Skeletal muscle can grow in three ways, by generation of new syncytial fibres, addition of nuclei from muscle stem cells to existing fibres or increase in cytoplasmic volume/nucleus. Evidence suggests the latter two processes contribute to exercise-induced growth. Fibre growth requires increase in sarcolemmal surface area and cytoplasmic volume at different rates. It has long been known that high-force exercise is a particularly effective growth stimulus, but how this stimulus is sensed and drives coordinated growth that is appropriately scaled across organelles remains a mystery.
Collapse
Affiliation(s)
- Michael Attwaters
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| |
Collapse
|
69
|
Grifell-Junyent M, Baum JF, Välimets S, Herrmann A, Paulusma CC, López-Marqués RL, Günther Pomorski T. CDC50A is required for aminophospholipid transport and cell fusion in mouse C2C12 myoblasts. J Cell Sci 2022; 135:jcs258649. [PMID: 34664668 PMCID: PMC10405909 DOI: 10.1242/jcs.258649] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/02/2021] [Indexed: 11/20/2022] Open
Abstract
Myoblast fusion is essential for the formation of multinucleated muscle fibers and is promoted by transient changes in the plasma membrane lipid distribution. However, little is known about the lipid transporters regulating these dynamic changes. Here, we show that proliferating myoblasts exhibit an aminophospholipid flippase activity that is downregulated during differentiation. Deletion of the P4-ATPase flippase subunit CDC50A (also known as TMEM30A) results in loss of the aminophospholipid flippase activity and compromises actin remodeling, RAC1 GTPase membrane targeting and cell fusion. In contrast, deletion of the P4-ATPase ATP11A affects aminophospholipid uptake without having a strong impact on cell fusion. Our results demonstrate that myoblast fusion depends on CDC50A and may involve multiple CDC50A-dependent P4-ATPases that help to regulate actin remodeling.
Collapse
Affiliation(s)
- Marta Grifell-Junyent
- Department of Molecular Biochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum, Germany
- Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Julia F. Baum
- Department of Molecular Biochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Silja Välimets
- Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Andreas Herrmann
- Institut für Biologie, Molekulare Biophysik, IRI Life Sciences, Humboldt-Universität zu Berlin, Invalidenstrasse 42, 10115 Berlin, Germany
| | - Coen C. Paulusma
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Rosa L. López-Marqués
- Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Thomas Günther Pomorski
- Department of Molecular Biochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum, Germany
- Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| |
Collapse
|
70
|
Palma-Vera SE, Reyer H, Langhammer M, Reinsch N, Derezanin L, Fickel J, Qanbari S, Weitzel JM, Franzenburg S, Hemmrich-Stanisak G, Schoen J. Genomic characterization of the world's longest selection experiment in mouse reveals the complexity of polygenic traits. BMC Biol 2022; 20:52. [PMID: 35189878 PMCID: PMC8862358 DOI: 10.1186/s12915-022-01248-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/07/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Long-term selection experiments are a powerful tool to understand the genetic background of complex traits. The longest of such experiments has been conducted in the Research Institute for Farm Animal Biology (FBN), generating extreme mouse lines with increased fertility, body mass, protein mass and endurance. For >140 generations, these lines have been maintained alongside an unselected control line, representing a valuable resource for understanding the genetic basis of polygenic traits. However, their history and genomes have not been reported in a comprehensive manner yet. Therefore, the aim of this study is to provide a summary of the breeding history and phenotypic traits of these lines along with their genomic characteristics. We further attempt to decipher the effects of the observed line-specific patterns of genetic variation on each of the selected traits. RESULTS Over the course of >140 generations, selection on the control line has given rise to two extremely fertile lines (>20 pups per litter each), two giant growth lines (one lean, one obese) and one long-distance running line. Whole genome sequencing analysis on 25 animals per line revealed line-specific patterns of genetic variation among lines, as well as high levels of homozygosity within lines. This high degree of distinctiveness results from the combined effects of long-term continuous selection, genetic drift, population bottleneck and isolation. Detection of line-specific patterns of genetic differentiation and structural variation revealed multiple candidate genes behind the improvement of the selected traits. CONCLUSIONS The genomes of the Dummerstorf trait-selected mouse lines display distinct patterns of genomic variation harbouring multiple trait-relevant genes. Low levels of within-line genetic diversity indicate that many of the beneficial alleles have arrived to fixation alongside with neutral alleles. This study represents the first step in deciphering the influence of selection and neutral evolutionary forces on the genomes of these extreme mouse lines and depicts the genetic complexity underlying polygenic traits.
Collapse
Affiliation(s)
- Sergio E Palma-Vera
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany.
| | - Henry Reyer
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Martina Langhammer
- Institute of Genetics and Biometry, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Norbert Reinsch
- Institute of Genetics and Biometry, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Lorena Derezanin
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- Department of Evolutionary Genetics, Research Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| | - Joerns Fickel
- Department of Evolutionary Genetics, Research Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
- University of Potsdam, Institute for Biochemistry and Biology, Potsdam, Germany
| | - Saber Qanbari
- Institute of Genetics and Biometry, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Joachim M Weitzel
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | | | | | - Jennifer Schoen
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- Department of Reproduction Biology, Research Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| |
Collapse
|
71
|
Suresh P, London E. Using cyclodextrin-induced lipid substitution to study membrane lipid and ordered membrane domain (raft) function in cells. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183774. [PMID: 34534531 PMCID: PMC9128603 DOI: 10.1016/j.bbamem.2021.183774] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 02/03/2023]
Abstract
Methods for efficient cyclodextrin-induced lipid exchange have been developed in our lab. These make it possible to almost completely replace the lipids in the outer leaflet of artificial membranes or the plasma membranes of living cells with exogenous lipids. Lipid replacement/substitution allows detailed studies of how lipid composition and asymmetry influence the structure and function of membrane domains and membrane proteins. In this review, we both summarize progress on cyclodextrin exchange in cells, mainly by the use of methyl-alpha cyclodextrin to exchange phospholipids and sphingolipids, and discuss the issues to consider when carrying out lipid exchange experiments upon cells. Issues that impact interpretation of lipid exchange are also discussed. This includes how overly naïve interpretation of how lipid exchange-induced changes in domain formation can impact protein function.
Collapse
|
72
|
Fine-Tuning of Piezo1 Expression and Activity Ensures Efficient Myoblast Fusion during Skeletal Myogenesis. Cells 2022; 11:cells11030393. [PMID: 35159201 PMCID: PMC8834081 DOI: 10.3390/cells11030393] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/22/2021] [Accepted: 01/19/2022] [Indexed: 01/12/2023] Open
Abstract
Mechanical stimuli, such as stretch and resistance training, are essential in regulating the growth and functioning of skeletal muscles. However, the molecular mechanisms involved in sensing mechanical stress during muscle formation remain unclear. Here, we investigated the role of the mechanosensitive ion channel Piezo1 during myogenic progression of both fast and slow muscle satellite cells. We found that Piezo1 level increases during myogenic differentiation and direct manipulation of Piezo1 in muscle stem cells alters the myogenic progression. Indeed, Piezo1 knockdown suppresses myoblast fusion, leading to smaller myotubes. Such an event is accompanied by significant downregulation of the fusogenic protein Myomaker. In parallel, while Piezo1 knockdown also lowers Ca2+ influx in response to stretch, Piezo1 activation increases Ca2+ influx in response to stretch and enhances myoblasts fusion. These findings may help understand molecular defects present in some muscle diseases. Our study shows that Piezo1 is essential for terminal muscle differentiation acting on myoblast fusion, suggesting that Piezo1 deregulation may have implications in muscle aging and degenerative diseases, including muscular dystrophies and neuromuscular disorders.
Collapse
|
73
|
Wang MJ, Zhu YC, Shi J. A crucial physiological role of Piezo1 channel in differentiation rather than proliferation during myogenesis. Acta Physiol (Oxf) 2021; 233:e13728. [PMID: 34492170 DOI: 10.1111/apha.13728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Ming Jie Wang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine Department of Physiology and Pathophysiology School of Basic Medical Sciences Fudan University Shanghai China
| | - Yi Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine Department of Physiology and Pathophysiology School of Basic Medical Sciences Fudan University Shanghai China
| | - Jian Shi
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine Department of Physiology and Pathophysiology School of Basic Medical Sciences Fudan University Shanghai China
- Leeds Institute of Cardiovascular and Metabolic Medicine School of Medicine University of Leeds Leeds UK
| |
Collapse
|
74
|
Bosutti A, Giniatullin A, Odnoshivkina Y, Giudice L, Malm T, Sciancalepore M, Giniatullin R, D'Andrea P, Lorenzon P, Bernareggi A. "Time window" effect of Yoda1-evoked Piezo1 channel activity during mouse skeletal muscle differentiation. Acta Physiol (Oxf) 2021; 233:e13702. [PMID: 34097801 PMCID: PMC9286833 DOI: 10.1111/apha.13702] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022]
Abstract
Aim Mechanosensitive Piezo1 ion channels emerged recently as important contributors to various vital functions including modulation of the blood supply to skeletal muscles. The specific Piezo1 channel agonist Yoda1 was shown to regulate the tone of blood vessels similarly to physical exercise. However, the direct role of Piezo1 channels in muscle function has been little studied so far. We therefore investigated the action of Yoda1 on the functional state of skeletal muscle precursors (satellite cells and myotubes) and on adult muscle fibres. Methods Immunostaining, electrophysiological intracellular recordings and Ca2+ imaging experiments were performed to localize and assess the effect of the chemical activation of Piezo1 channels with Yoda1, on myogenic precursors, adult myofibres and at the adult neuromuscular junction. Results Piezo1 channels were detected by immunostaining in satellite cells (SCs) and myotubes as well as in adult myofibres. In the skeletal muscle precursors, Yoda1 treatment stimulated the differentiation and cell fusion rather than the proliferation of SCs. Moreover, in myotubes, Yoda1 induced significant [Ca2+]i transients, without detectable [Ca2+]i response in adult myofibres. Furthermore, although expression of Piezo1 channels was detected around the muscle endplate region, Yoda1 application did not alter either the nerve‐evoked or spontaneous synaptic activity or muscle contractions in adult myofibres. Conclusion Our data indicate that the chemical activation of Piezo1 channels specifically enhances the differentiation of skeletal muscle precursors, suggesting a possible new strategy to promote muscle regeneration.
Collapse
Affiliation(s)
| | - Arthur Giniatullin
- Department of Physiology Kazan State Medical University Kazan Russia
- Laboratory of Biophysics of Synaptic Processes Kazan Institute of Biochemistry and BiophysicsFederal Research Center “Kazan Scientific Center of RAS” Kazan Russia
| | | | - Luca Giudice
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Marina Sciancalepore
- Department of Life Sciences University of Trieste Trieste Italy
- B.R.A.I.N., University of Trieste Centre for Neuroscience Trieste Italy
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
- Institute of Fundamental Medicine and Biology Federal University Kazan Russia
| | - Paola D'Andrea
- Department of Life Sciences University of Trieste Trieste Italy
| | - Paola Lorenzon
- Department of Life Sciences University of Trieste Trieste Italy
- B.R.A.I.N., University of Trieste Centre for Neuroscience Trieste Italy
| | - Annalisa Bernareggi
- Department of Life Sciences University of Trieste Trieste Italy
- B.R.A.I.N., University of Trieste Centre for Neuroscience Trieste Italy
| |
Collapse
|
75
|
Orsini EM, Perelas A, Southern BD, Grove LM, Olman MA, Scheraga RG. Stretching the Function of Innate Immune Cells. Front Immunol 2021; 12:767319. [PMID: 34795674 PMCID: PMC8593101 DOI: 10.3389/fimmu.2021.767319] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
The importance of innate immune cells to sense and respond to their physical environment is becoming increasingly recognized. Innate immune cells (e.g. macrophages and neutrophils) are able to receive mechanical signals through several mechanisms. In this review, we discuss the role of mechanosensitive ion channels, such as Piezo1 and transient receptor potential vanilloid 4 (TRPV4), and cell adhesion molecules, such as integrins, selectins, and cadherins in biology and human disease. Furthermore, we explain that these mechanical stimuli activate intracellular signaling pathways, such as MAPK (p38, JNK), YAP/TAZ, EDN1, NF-kB, and HIF-1α, to induce protein conformation changes and modulate gene expression to drive cellular function. Understanding the mechanisms by which immune cells interpret mechanosensitive information presents potential targets to treat human disease. Important areas of future study in this area include autoimmune, allergic, infectious, and malignant conditions.
Collapse
Affiliation(s)
- Erica M Orsini
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Apostolos Perelas
- Department of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA, United States
| | - Brian D Southern
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Lisa M Grove
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Mitchell A Olman
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Rachel G Scheraga
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
76
|
Sun KX, Jiang XY, Li X, Su YJ, Wang JL, Zhang L, Yang YM, Zhu XJ. Deletion of phosphatidylserine flippase β-subunit Tmem30a in satellite cells leads to delayed skeletal muscle regeneration. Zool Res 2021; 42:650-659. [PMID: 34472226 PMCID: PMC8455468 DOI: 10.24272/j.issn.2095-8137.2021.195] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Phosphatidylserine (PS) is distributed asymmetrically in the plasma membrane of eukaryotic cells. Phosphatidylserine flippase (P4-ATPase) transports PS from the outer leaflet of the lipid bilayer to the inner leaflet of the membrane to maintain PS asymmetry. The β subunit TMEM30A is indispensable for transport and proper function of P4-ATPase. Previous studies have shown that the ATP11A and TMEM30A complex is the molecular switch for myotube formation. However, the role of Tmem30a in skeletal muscle regeneration remains elusive. In the current study, Tmem30a was highly expressed in the tibialis anterior (TA) muscles of dystrophin-null (mdx) mice and BaCl2-induced muscle injury model mice. We generated a satellite cell (SC)-specific Tmem30a conditional knockout (cKO) mouse model to investigate the role of Tmem30a in skeletal muscle regeneration. The regenerative ability of cKO mice was evaluated by analyzing the number and diameter of regenerated SCs after the TA muscles were injured by BaCl2-injection. Compared to the control mice, the cKO mice showed decreased Pax7+ and MYH3+ SCs, indicating diminished SC proliferation, and decreased expression of muscular regulatory factors (MYOD and MYOG), suggesting impaired myoblast proliferation in skeletal muscle regeneration. Taken together, these results demonstrate the essential role of Tmem30a in skeletal muscle regeneration.
Collapse
Affiliation(s)
- Kuan-Xiang Sun
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072 China
| | - Xiao-Yan Jiang
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xiao Li
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Yu-Jing Su
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Ju-Lin Wang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Lin Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Ye-Ming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xian-Jun Zhu
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072 China.,Department of Ophthalmology, First People's Hospital of Shangqiu, Shangqiu, Henan 476000, China.,Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China. E-mail:
| |
Collapse
|
77
|
Li JV, Ng CA, Cheng D, Zhou Z, Yao M, Guo Y, Yu ZY, Ramaswamy Y, Ju LA, Kuchel PW, Feneley MP, Fatkin D, Cox CD. Modified N-linked glycosylation status predicts trafficking defective human Piezo1 channel mutations. Commun Biol 2021; 4:1038. [PMID: 34489534 PMCID: PMC8421374 DOI: 10.1038/s42003-021-02528-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023] Open
Abstract
Mechanosensitive channels are integral membrane proteins that sense mechanical stimuli. Like most plasma membrane ion channel proteins they must pass through biosynthetic quality control in the endoplasmic reticulum that results in them reaching their destination at the plasma membrane. Here we show that N-linked glycosylation of two highly conserved asparagine residues in the 'cap' region of mechanosensitive Piezo1 channels are necessary for the mature protein to reach the plasma membrane. Both mutation of these asparagines (N2294Q/N2331Q) and treatment with an enzyme that hydrolyses N-linked oligosaccharides (PNGaseF) eliminates the fully glycosylated mature Piezo1 protein. The N-glycans in the cap are a pre-requisite for N-glycosylation in the 'propeller' regions, which are present in loops that are essential for mechanotransduction. Importantly, trafficking-defective Piezo1 variants linked to generalized lymphatic dysplasia and bicuspid aortic valve display reduced fully N-glycosylated Piezo1 protein. Thus the N-linked glycosylation status in vitro correlates with efficient membrane trafficking and will aid in determining the functional impact of Piezo1 variants of unknown significance.
Collapse
Affiliation(s)
- Jinyuan Vero Li
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Chai-Ann Ng
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Delfine Cheng
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Zijing Zhou
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Mingxi Yao
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Yang Guo
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Ze-Yan Yu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Camperdown, NSW, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Camperdown, NSW, Australia
| | - Philip W Kuchel
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Michael P Feneley
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
- Department of Cardiology, St Vincent's Hospital, Sydney, Australia
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
78
|
Günay KA, Silver JS, Chang TL, Bednarski OJ, Bannister KL, Rogowski CJ, Olwin BB, Anseth KS. Myoblast mechanotransduction and myotube morphology is dependent on BAG3 regulation of YAP and TAZ. Biomaterials 2021; 277:121097. [PMID: 34481290 DOI: 10.1016/j.biomaterials.2021.121097] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/26/2021] [Accepted: 08/24/2021] [Indexed: 11/30/2022]
Abstract
Skeletal muscle tissue is mechanically dynamic with changes in stiffness influencing function, maintenance, and regeneration. We modeled skeletal muscle mechanical changes in culture with dynamically stiffening hydrogels demonstrating that the chaperone protein BAG3 transduces matrix stiffness by redistributing YAP and TAZ subcellular localization in muscle progenitor cells. BAG3 depletion increases cytoplasmic retention of YAP and TAZ, desensitizing myoblasts to changes in hydrogel elastic moduli. Upon differentiation, muscle progenitors depleted of BAG3 formed enlarged, round myotubes lacking the typical cylindrical morphology. The aberrant morphology is dependent on YAP/TAZ signaling, which was sequestered in the cytoplasm in BAG3-depleted myotubes but predominately nuclear in cylindrical myotubes of control cells. Control progenitor cells induced to differentiate on soft (E' = 4 and 12 kPa) hydrogels formed circular myotubes similar to those observed in BAG3-depleted cells. Inhibition of the Hippo pathway partially restored myotube morphologies, permitting nuclear translocation of YAP and TAZ in BAG3-depleted myogenic progenitors. Thus, BAG3 is a critical mediator of dynamic stiffness changes in muscle tissue, coupling mechanical alterations to intracellular signals and inducing changes in gene expression that influence muscle progenitor cell morphology and differentiation.
Collapse
Affiliation(s)
- K Arda Günay
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Jason S Silver
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA; Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA; Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tze-Ling Chang
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Olivia J Bednarski
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Kendra L Bannister
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Cameron J Rogowski
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Bradley B Olwin
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA; Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA.
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA.
| |
Collapse
|
79
|
Cioffi F, Adam RHI, Bansal R, Broersen K. A Review of Oxidative Stress Products and Related Genes in Early Alzheimer's Disease. J Alzheimers Dis 2021; 83:977-1001. [PMID: 34420962 PMCID: PMC8543250 DOI: 10.3233/jad-210497] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxidative stress is associated with the progression of Alzheimer’s disease (AD). Reactive oxygen species can modify lipids, DNA, RNA, and proteins in the brain. The products of their peroxidation and oxidation are readily detectable at incipient stages of disease. Based on these oxidation products, various biomarker-based strategies have been developed to identify oxidative stress levels in AD. Known oxidative stress-related biomarkers include lipid peroxidation products F2-isoprostanes, as well as malondialdehyde and 4-hydroxynonenal which both conjugate to specific amino acids to modify proteins, and DNA or RNA oxidation products 8-hydroxy-2’-deoxyguanosine (8-OHdG) and 8-hydroxyguanosine (8-OHG), respectively. The inducible enzyme heme oxygenase type 1 (HO-1) is found to be upregulated in response to oxidative stress-related events in the AD brain. While these global biomarkers for oxidative stress are associated with early-stage AD, they generally poorly differentiate from other neurodegenerative disorders that also coincide with oxidative stress. Redox proteomics approaches provided specificity of oxidative stress-associated biomarkers to AD pathology by the identification of oxidatively damaged pathology-specific proteins. In this review, we discuss the potential combined diagnostic value of these reported biomarkers in the context of AD and discuss eight oxidative stress-related mRNA biomarkers in AD that we newly identified using a transcriptomics approach. We review these genes in the context of their reported involvement in oxidative stress regulation and specificity for AD. Further research is warranted to establish the protein levels and their functionalities as well as the molecular mechanisms by which these potential biomarkers are involved in regulation of oxidative stress levels and their potential for determination of oxidative stress and disease status of AD patients.
Collapse
Affiliation(s)
- Federica Cioffi
- Department of Nanobiophysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Rayan Hassan Ibrahim Adam
- Department of Nanobiophysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Department of Medical Cell Biophysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.,Department of Pharmacokinetics, Toxicology, and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Kerensa Broersen
- Department of Applied Stem Cell Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
80
|
Deisl C, Hilgemann DW, Syeda R, Fine M. TMEM16F and dynamins control expansive plasma membrane reservoirs. Nat Commun 2021; 12:4990. [PMID: 34404808 PMCID: PMC8371123 DOI: 10.1038/s41467-021-25286-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/29/2021] [Indexed: 11/09/2022] Open
Abstract
Cells can expand their plasma membrane laterally by unfolding membrane undulations and by exocytosis. Here, we describe a third mechanism involving invaginations held shut by the membrane adapter, dynamin. Compartments open when Ca activates the lipid scramblase, TMEM16F, anionic phospholipids escape from the cytoplasmic monolayer in exchange for neutral lipids, and dynamins relax. Deletion of TMEM16F or dynamins blocks expansion, with loss of dynamin expression generating a maximally expanded basal plasma membrane state. Re-expression of dynamin2 or its GTPase-inactivated mutant, but not a lipid binding mutant, regenerates reserve compartments and rescues expansion. Dynamin2-GFP fusion proteins form punctae that rapidly dissipate from these compartments during TMEM16F activation. Newly exposed compartments extend deeply into the cytoplasm, lack numerous organellar markers, and remain closure-competent for many seconds. Without Ca, compartments open slowly when dynamins are sequestered by cytoplasmic dynamin antibodies or when scrambling is mimicked by neutralizing anionic phospholipids and supplementing neutral lipids. Activation of Ca-permeable mechanosensitive channels via cell swelling or channel agonists opens the compartments in parallel with phospholipid scrambling. Thus, dynamins and TMEM16F control large plasma membrane reserves that open in response to lateral membrane stress and Ca influx.
Collapse
Affiliation(s)
- Christine Deisl
- University of Texas Southwestern Medical Center, Department of Physiology, Dallas, TX, USA
| | - Donald W Hilgemann
- University of Texas Southwestern Medical Center, Department of Physiology, Dallas, TX, USA.
| | - Ruhma Syeda
- University of Texas Southwestern Medical Center, Department of Neuroscience, Dallas, TX, USA
| | - Michael Fine
- University of Texas Southwestern Medical Center, Department of Physiology, Dallas, TX, USA.
- University of Texas Southwestern Medical Center, Department of Molecular Genetics, Dallas, TX, USA.
| |
Collapse
|
81
|
Ristovski M, Farhat D, Bancud SEM, Lee JY. Lipid Transporters Beam Signals from Cell Membranes. MEMBRANES 2021; 11:562. [PMID: 34436325 PMCID: PMC8399137 DOI: 10.3390/membranes11080562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022]
Abstract
Lipid composition in cellular membranes plays an important role in maintaining the structural integrity of cells and in regulating cellular signaling that controls functions of both membrane-anchored and cytoplasmic proteins. ATP-dependent ABC and P4-ATPase lipid transporters, two integral membrane proteins, are known to contribute to lipid translocation across the lipid bilayers on the cellular membranes. In this review, we will highlight current knowledge about the role of cholesterol and phospholipids of cellular membranes in regulating cell signaling and how lipid transporters participate this process.
Collapse
Affiliation(s)
- Miliça Ristovski
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.R.); (D.F.); (S.E.M.B.)
- Translational and Molecular Medicine Program, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Danny Farhat
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.R.); (D.F.); (S.E.M.B.)
- Biomedical Sciences Program, Faculty of Science, University of Ottawa, Ottawa, ON K1H 6N5, Canada
| | - Shelly Ellaine M. Bancud
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.R.); (D.F.); (S.E.M.B.)
- Translational and Molecular Medicine Program, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jyh-Yeuan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.R.); (D.F.); (S.E.M.B.)
| |
Collapse
|
82
|
Zhang X, Zhang J, Wu Y, Nan B, Huang Q, Du X, Tian M, Liu L, Xin Y, Li Y, Duan J, Chen R, Sun Z, Shen H. Dynamic recovery after acute single fine particulate matter exposure in male mice: Effect on lipid deregulation and cardiovascular alterations. JOURNAL OF HAZARDOUS MATERIALS 2021; 414:125504. [PMID: 33652219 DOI: 10.1016/j.jhazmat.2021.125504] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 06/12/2023]
Abstract
Many studies have linked airborne fine particulate matter (PM2.5) exposure to cardiovascular diseases. We performed a time-series analysis to investigate whether the disruption of lipid metabolism recovered or lasted after acute PM2.5 exposure in mice. Targeted lipidomic analysis showed that four major plasma membrane phospholipids along with cholesterol esters (CE) were significantly altered on 7th post-exposure day (PED7), and the alteration reached a peak on PED14. On PED21, the phosphatidylcholine (PC) decrease was more marked than on PED14, and its resurgence was indirectly linked to triglyceride (TG) increase. Homocysteine (HCY), lactate dehydrogenase (LDH), and α-hydroxybutyrate dehydrogenase (α-HBDH) levels increased but glucose levels decreased markedly in a dose- and time-dependent manner throughout the experimental period. Network analysis showed that the lasting lipid deregulation on PED21 correlated to myocardial markers and glucose interruption, during which high-density lipoprotein cholesterol (HDL-C) decreased. The present data implied that the constructional membrane lipids were initially interrupted by PM2.5, and the subsequent rehabilitation resulted in the deregulation of storage lipids; the parallel myocardial and glucose effects may be enhanced by the lasting HDL-C lipid deregulation on PED21. These myocardial and lipidomic events were early indicators of cardiovascular risk, resulting from subsequent exposure to and accumulation of PM2.5.
Collapse
Affiliation(s)
- Xi Zhang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jie Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Yan Wu
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, PR China
| | - Bingru Nan
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Qingyu Huang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Xiaoyan Du
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Meiping Tian
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Liangpo Liu
- School of Public Health, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yuntian Xin
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yanbo Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Rui Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Heqing Shen
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, PR China.
| |
Collapse
|
83
|
Shah V, Patel S, Shah J. Emerging Role of Piezo Ion Channels in Cardiovascular Development. Dev Dyn 2021; 251:276-286. [PMID: 34255896 DOI: 10.1002/dvdy.401] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/12/2021] [Accepted: 07/09/2021] [Indexed: 12/23/2022] Open
Abstract
Mechanical cues are crucial for vascular development and the proper differentiation of various cell types. Piezo1 and Piezo2 are mechanically activated cationic channels expressed in various cell types, especially in vascular smooth muscle and endothelial cells. It is present as a transmembrane homotrimeric complex, regulating calcium influx. Local blood flow associated shear stress, in addition to blood pressure associated cell membrane stretching are key Piezo channel activators. There is rising proof, showcasing Piezo channels significance in myocytes, cardiac fibroblast, vascular tone maintenance, atherosclerosis, hypertension, NO generation, and baroreceptor reflex. Here, we review the role of Piezo channels in cardiovascular development and its associated clinical disorders. Also, emphasizing on Piezo channel modulators which might lead to novel therapies for cardiovascular diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Vandit Shah
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Sandip Patel
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Jigna Shah
- Department of Pharmacology, Institute of Pharmacy Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
84
|
Kumari P, Faraone A, Kelley EG, Benedetto A. Stiffening Effect of the [Bmim][Cl] Ionic Liquid on the Bending Dynamics of DMPC Lipid Vesicles. J Phys Chem B 2021; 125:7241-7250. [PMID: 34169716 PMCID: PMC8279542 DOI: 10.1021/acs.jpcb.1c01347] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The elastic properties of the cellular lipid membrane play a crucial role for life. Their alteration can lead to cell malfunction, and in turn, being able to control them holds the promise of effective therapeutic and diagnostic approaches. In this context, due to their proven strong interaction with lipid bilayers, ionic liquids (ILs)-a vast class of organic electrolytes-may play an important role. This work focuses on the effect of the model imidazolium-IL [bmim][Cl] on the bending modulus of DMPC lipid vesicles, a basic model of cellular lipid membranes. Here, by combining small-angle neutron scattering and neutron spin-echo spectroscopy, we show that the IL, dispersed at low concentrations at the bilayer-water interface, (i) diffuses into the lipid region, accounting for five IL-cations for every 11 lipids, and (ii) causes an increase of the lipid bilayer bending modulus, up to 60% compared to the neat lipid bilayer at 40 °C.
Collapse
Affiliation(s)
- Pallavi Kumari
- Department of Sciences, University of Roma Tre, 00146 Rome, Italy.,School of Physics and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Antonio Faraone
- NIST Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Elizabeth G Kelley
- NIST Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Antonio Benedetto
- Department of Sciences, University of Roma Tre, 00146 Rome, Italy.,School of Physics and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland.,Laboratory for Neutron Scattering, Paul Scherrer Institute, 5232 Villigen, Switzerland
| |
Collapse
|
85
|
Dumitru AC, Stommen A, Koehler M, Cloos AS, Yang J, Leclercqz A, Tyteca D, Alsteens D. Probing PIEZO1 Localization upon Activation Using High-Resolution Atomic Force and Confocal Microscopy. NANO LETTERS 2021; 21:4950-4958. [PMID: 34125553 DOI: 10.1021/acs.nanolett.1c00599] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
PIEZO1 ion channels are activated by mechanical stimuli, triggering intracellular chemical signals. Recent structural studies suggest that plasma membrane tension or local curvature changes modulate PIEZO1 channel gating and activation. However, whether PIEZO1 localization is governed by tension gradients or long-range mechanical perturbations across the cells is still unclear. Here, we probe the nanoscale localization of PIEZO1 on red blood cells (RBCs) at high resolution (∼30 nm), and we report for the first time the existence of submicrometric PIEZO1 clusters in native conditions. Upon interaction with Yoda1, an allosteric modulator, PIEZO1 clusters increase in abundance in regions of higher membrane tension and lower curvature. We further show that PIEZO1 ion channels interact with the spectrin cytoskeleton in both resting and activated states. Our results point toward a strong interplay between plasma membrane tension gradients, curvature, and cytoskeleton association of PIEZO1.
Collapse
Affiliation(s)
- Andra C Dumitru
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Amaury Stommen
- de Duve Institute, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Anne-Sophie Cloos
- de Duve Institute, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Jinsung Yang
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Arnaud Leclercqz
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Donatienne Tyteca
- de Duve Institute, Université Catholique de Louvain, Brussels 1200, Belgium
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| |
Collapse
|
86
|
Jiang W, Lin YC, Luo YL. Mechanical properties of anionic asymmetric bilayers from atomistic simulations. J Chem Phys 2021; 154:224701. [PMID: 34241213 PMCID: PMC8189722 DOI: 10.1063/5.0048232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/12/2021] [Indexed: 11/14/2022] Open
Abstract
Mechanotransduction, the biological response to mechanical stress, is often initiated by activation of mechanosensitive (MS) proteins upon mechanically induced deformations of the cell membrane. A current challenge in fully understanding this process is in predicting how lipid bilayers deform upon the application of mechanical stress. In this context, it is now well established that anionic lipids influence the function of many proteins. Here, we test the hypothesis that anionic lipids could indirectly modulate MS proteins by alteration of the lipid bilayer mechanical properties. Using all-atom molecular dynamics simulations, we computed the bilayer bending rigidity (KC), the area compressibility (KA), and the surface shear viscosity (ηm) of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (PC) lipid bilayers with and without phosphatidylserine (PS) or phosphatidylinositol bisphosphate (PIP2) at physiological concentrations in the lower leaflet. Tensionless leaflets were first checked for each asymmetric bilayer model, and a formula for embedding an asymmetric channel in an asymmetric bilayer is proposed. Results from two different sized bilayers show consistently that the addition of 20% surface charge in the lower leaflet of the PC bilayer with PIP2 has minimal impact on its mechanical properties, while PS reduced the bilayer bending rigidity by 22%. As a comparison, supplementing the PIP2-enriched PC membrane with 30% cholesterol, a known rigidifying steroid lipid, produces a significant increase in all three mechanical constants. Analysis of pairwise splay moduli suggests that the effect of anionic lipids on bilayer bending rigidity largely depends on the number of anionic lipid pairs formed during simulations. The potential implication of bilayer bending rigidity is discussed in the framework of MS piezo channels.
Collapse
Affiliation(s)
- Wenjuan Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California 91766, USA
| | - Yi-Chun Lin
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California 91766, USA
| | - Yun Lyna Luo
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California 91766, USA
| |
Collapse
|
87
|
Shiomi A, Nagao K, Yokota N, Tsuchiya M, Kato U, Juni N, Hara Y, Mori MX, Mori Y, Ui-Tei K, Murate M, Kobayashi T, Nishino Y, Miyazawa A, Yamamoto A, Suzuki R, Kaufmann S, Tanaka M, Tatsumi K, Nakabe K, Shintaku H, Yesylevsky S, Bogdanov M, Umeda M. Extreme deformability of insect cell membranes is governed by phospholipid scrambling. Cell Rep 2021; 35:109219. [PMID: 34107250 DOI: 10.1016/j.celrep.2021.109219] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 04/02/2021] [Accepted: 05/13/2021] [Indexed: 10/21/2022] Open
Abstract
Organization of dynamic cellular structure is crucial for a variety of cellular functions. In this study, we report that Drosophila and Aedes have highly elastic cell membranes with extremely low membrane tension and high resistance to mechanical stress. In contrast to other eukaryotic cells, phospholipids are symmetrically distributed between the bilayer leaflets of the insect plasma membrane, where phospholipid scramblase (XKR) that disrupts the lipid asymmetry is constitutively active. We also demonstrate that XKR-facilitated phospholipid scrambling promotes the deformability of cell membranes by regulating both actin cortex dynamics and mechanical properties of the phospholipid bilayer. Moreover, XKR-mediated construction of elastic cell membranes is essential for hemocyte circulation in the Drosophila cardiovascular system. Deformation of mammalian cells is also enhanced by the expression of Aedes XKR, and thus phospholipid scrambling may contribute to formation of highly deformable cell membranes in a variety of living eukaryotic cells.
Collapse
Affiliation(s)
- Akifumi Shiomi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan
| | - Kohjiro Nagao
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan.
| | - Nobuhiro Yokota
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan
| | - Masaki Tsuchiya
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan
| | - Utako Kato
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan
| | - Naoto Juni
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan
| | - Yuji Hara
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan
| | - Masayuki X Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan
| | - Kumiko Ui-Tei
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Motohide Murate
- UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, 74 Route du Rhin, 67401 Illkirch, France
| | - Toshihide Kobayashi
- UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, 74 Route du Rhin, 67401 Illkirch, France
| | - Yuri Nishino
- Graduate School of Life Science, University of Hyogo, Kouto, Kamigori-cho, Ako-gun, Hyogo 678-1297, Japan
| | - Atsuo Miyazawa
- Graduate School of Life Science, University of Hyogo, Kouto, Kamigori-cho, Ako-gun, Hyogo 678-1297, Japan
| | - Akihisa Yamamoto
- Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan
| | - Ryo Suzuki
- Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan
| | - Stefan Kaufmann
- Physical Chemistry of Biosystems, Institute of Physical Chemistry, University of Heidelberg, 69120 Heidelberg, Germany
| | - Motomu Tanaka
- Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan; Physical Chemistry of Biosystems, Institute of Physical Chemistry, University of Heidelberg, 69120 Heidelberg, Germany
| | - Kazuya Tatsumi
- Department of Mechanical Engineering and Science, Kyoto University, Katsura, Kyoto 615-8540, Japan
| | - Kazuyoshi Nakabe
- Department of Mechanical Engineering and Science, Kyoto University, Katsura, Kyoto 615-8540, Japan
| | - Hirofumi Shintaku
- Microfluidics RIKEN Hakubi Research Team, RIKEN Cluster for Pioneering Research, Wako, Saitama 351-0198, Japan
| | - Semen Yesylevsky
- Laboratoire Chrono Environnement UMR CNRS 6249, Université de Bourgogne Franche-Comté, 16 Route de Gray, 25030 Besançon Cedex, France; Department of Physics of Biological Systems, Institute of Physics of the National Academy of Sciences of Ukraine, Prospect Nauky 46, 03680 Kyiv, Ukraine
| | - Mikhail Bogdanov
- Department of Biochemistry & Molecular Biology, University of Texas Health Science Center at Houston, McGovern Medical School, 6431 Fannin, Houston, TX 77030, USA
| | - Masato Umeda
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Kyoto 615-8510, Japan.
| |
Collapse
|
88
|
Mioka T, Guo T, Wang S, Tsuji T, Kishimoto T, Fujimoto T, Tanaka K. Characterization of micron-scale protein-depleted plasma membrane domains in phosphatidylserine-deficient yeast cells. J Cell Sci 2021; 135:261783. [PMID: 34000034 DOI: 10.1242/jcs.256529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/16/2021] [Indexed: 12/30/2022] Open
Abstract
Membrane phase separation to form micron-scale domains of lipids and proteins occurs in artificial membranes; however, a similar large-scale phase separation has not been reported in the plasma membrane of the living cells. We show here that a stable micron-scale protein-depleted region is generated in the plasma membrane of yeast mutants lacking phosphatidylserine at high temperatures. We named this region the 'void zone'. Transmembrane proteins and certain peripheral membrane proteins and phospholipids are excluded from the void zone. The void zone is rich in ergosterol, and requires ergosterol and sphingolipids for its formation. Such properties are also found in the cholesterol-enriched domains of phase-separated artificial membranes, but the void zone is a novel membrane domain that requires energy and various cellular functions for its formation. The formation of the void zone indicates that the plasma membrane in living cells has the potential to undergo phase separation with certain lipid compositions. We also found that void zones were frequently in contact with vacuoles, in which a membrane domain was also formed at the contact site.
Collapse
Affiliation(s)
- Tetsuo Mioka
- Division of Molecular Interaction, Institute for Genetic Medicine, Hokkaido University Graduate School of Life Science, Sapporo, Hokkaido 060-0815, Japan
| | - Tian Guo
- Division of Molecular Interaction, Institute for Genetic Medicine, Hokkaido University Graduate School of Life Science, Sapporo, Hokkaido 060-0815, Japan
| | - Shiyao Wang
- Division of Molecular Interaction, Institute for Genetic Medicine, Hokkaido University Graduate School of Life Science, Sapporo, Hokkaido 060-0815, Japan
| | - Takuma Tsuji
- Laboratory of Molecular Cell Biology, Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takuma Kishimoto
- Division of Molecular Interaction, Institute for Genetic Medicine, Hokkaido University Graduate School of Life Science, Sapporo, Hokkaido 060-0815, Japan
| | - Toyoshi Fujimoto
- Laboratory of Molecular Cell Biology, Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Kazuma Tanaka
- Division of Molecular Interaction, Institute for Genetic Medicine, Hokkaido University Graduate School of Life Science, Sapporo, Hokkaido 060-0815, Japan
| |
Collapse
|
89
|
Nakao H, Kimura Y, Sakai A, Ikeda K, Nakano M. Development of membrane-insertable lipid scrambling peptides: A time-resolved small-angle neutron scattering study. STRUCTURAL DYNAMICS (MELVILLE, N.Y.) 2021; 8:024301. [PMID: 33758768 PMCID: PMC7980860 DOI: 10.1063/4.0000045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/26/2021] [Indexed: 05/14/2023]
Abstract
Phospholipid transbilayer movement (flip-flop) in the plasma membrane is regulated by membrane proteins to maintain cell homeostasis and interact with other cells. The promotion of flip-flop by phospholipid scramblases causes the loss of membrane lipid asymmetry, which is involved in apoptosis, blood coagulation, and viral infection. Therefore, compounds that can artificially control flip-flop in the plasma membrane are of biological and medical interest. Here, we have developed lipid scrambling transmembrane peptides that can be inserted into the membrane. Time-resolved small-angle neutron scattering measurements revealed that the addition of peptides containing a glutamine residue at the center of the hydrophobic sequence to lipid vesicles induces the flip-flop of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine. Peptides without the glutamine residue had no effect on the flip-flop. Because the glutamine-containing peptides exhibited scramblase activity in monomeric form, the polar glutamine residue would be exposed to the hydrocarbon region of the membrane, perturbing the membrane and promoting the lipid flip-flop. These scrambling peptides would be valuable tools to regulate lipid flip-flop in the plasma membrane.
Collapse
Affiliation(s)
| | | | | | | | - Minoru Nakano
- Author to whom correspondence should be addressed: . Tel.: +81 76 434 7565. Fax: +81 76 434 7568
| |
Collapse
|
90
|
The transport mechanism of P4 ATPase lipid flippases. Biochem J 2021; 477:3769-3790. [PMID: 33045059 DOI: 10.1042/bcj20200249] [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] [Received: 07/02/2020] [Revised: 09/02/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022]
Abstract
P4 ATPase lipid flippases are ATP-driven transporters that translocate specific lipids from the exoplasmic to the cytosolic leaflet of biological membranes, thus establishing a lipid gradient between the two leaflets that is essential for many cellular processes. While substrate specificity, subcellular and tissue-specific expression, and physiological functions have been assigned to a number of these transporters in several organisms, the mechanism of lipid transport has been a topic of intense debate in the field. The recent publication of a series of structural models based on X-ray crystallography and cryo-EM studies has provided the first glimpse into how P4 ATPases have adapted the transport mechanism used by the cation-pumping family members to accommodate a substrate that is at least an order of magnitude larger than cations.
Collapse
|
91
|
Flagging fusion: Phosphatidylserine signaling in cell-cell fusion. J Biol Chem 2021; 296:100411. [PMID: 33581114 PMCID: PMC8005811 DOI: 10.1016/j.jbc.2021.100411] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Formations of myofibers, osteoclasts, syncytiotrophoblasts, and fertilized zygotes share a common step, cell–cell fusion. Recent years have brought about considerable progress in identifying some of the proteins involved in these and other cell-fusion processes. However, even for the best-characterized cell fusions, we still do not know the mechanisms that regulate the timing of cell-fusion events. Are they fully controlled by the expression of fusogenic proteins or do they also depend on some triggering signal that activates these proteins? The latter scenario would be analogous to the mechanisms that control the timing of exocytosis initiated by Ca2+ influx and virus-cell fusion initiated by low pH- or receptor interaction. Diverse cell fusions are accompanied by the nonapoptotic exposure of phosphatidylserine at the surface of fusing cells. Here we review data on the dependence of membrane remodeling in cell fusion on phosphatidylserine and phosphatidylserine-recognizing proteins and discuss the hypothesis that cell surface phosphatidylserine serves as a conserved “fuse me” signal regulating the time and place of cell-fusion processes.
Collapse
|
92
|
Enhanced Ca 2+ influx in mechanically distorted erythrocytes measured with 19F nuclear magnetic resonance spectroscopy. Sci Rep 2021; 11:3749. [PMID: 33580124 PMCID: PMC7881017 DOI: 10.1038/s41598-021-83044-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/19/2021] [Indexed: 01/30/2023] Open
Abstract
We present the first direct nuclear magnetic resonance (NMR) evidence of enhanced entry of Ca2+ ions into human erythrocytes (red blood cells; RBCs), when these cells are mechanically distorted. For this we loaded the RBCs with the fluorinated Ca2+ chelator, 1,2-bis(2-amino-5-fluorophenoxy)ethane-N,N,N',N'-tetraacetic acid (5FBAPTA), and recorded 19F NMR spectra. The RBCs were suspended in gelatin gel in a special stretching/compression apparatus. The 5FBAPTA was loaded into the cells as the tetraacetoxymethyl ester; and 13C NMR spectroscopy with [1,6-13C]D-glucose as substrate showed active glycolysis albeit at a reduced rate in cell suspensions and gels. The enhancement of Ca2+ influx is concluded to be via the mechanosensitive cation channel Piezo1. The increased rate of influx brought about by the activator of Piezo1, 2-[5-[[(2,6-dichlorophenyl)methyl]thio]-1,3,4-thiadiazol-2-yl]-pyrazine (Yoda1) supported this conclusion; while the specificity of the cation-sensing by 5FBAPTA was confirmed by using the Ca2+ ionophore, A23187.
Collapse
|
93
|
Jiang W, Del Rosario JS, Botello-Smith W, Zhao S, Lin YC, Zhang H, Lacroix J, Rohacs T, Luo YL. Crowding-induced opening of the mechanosensitive Piezo1 channel in silico. Commun Biol 2021; 4:84. [PMID: 33469156 PMCID: PMC7815867 DOI: 10.1038/s42003-020-01600-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
Mechanosensitive Piezo1 channels are essential mechanotransduction proteins in eukaryotes. Their curved transmembrane domains, called arms, create a convex membrane deformation, or footprint, which is predicted to flatten in response to increased membrane tension. Here, using a hyperbolic tangent model, we show that, due to the intrinsic bending rigidity of the membrane, the overlap of neighboring Piezo1 footprints produces a flattening of the Piezo1 footprints and arms. Multiple all-atom molecular dynamics simulations of Piezo1 further reveal that this tension-independent flattening is accompanied by gating motions that open an activation gate in the pore. This open state recapitulates experimentally obtained ionic selectivity, unitary conductance, and mutant phenotypes. Tracking ion permeation along the open pore reveals the presence of intracellular and extracellular fenestrations acting as cation-selective sites. Simulations also reveal multiple potential binding sites for phosphatidylinositol 4,5-bisphosphate. We propose that the overlap of Piezo channel footprints may act as a cooperative mechanism to regulate channel activity.
Collapse
Affiliation(s)
- Wenjuan Jiang
- College of Pharmacy, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - John Smith Del Rosario
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Wesley Botello-Smith
- College of Pharmacy, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Siyuan Zhao
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Yi-Chun Lin
- College of Pharmacy, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Han Zhang
- College of Pharmacy, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Jérôme Lacroix
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, 91766, USA.
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA.
| | - Yun Lyna Luo
- College of Pharmacy, Western University of Health Sciences, Pomona, CA, 91766, USA.
| |
Collapse
|
94
|
Winterstein LM, Kukovetz K, Hansen UP, Schroeder I, Van Etten JL, Moroni A, Thiel G, Rauh O. Distinct lipid bilayer compositions have general and protein-specific effects on K+ channel function. J Gen Physiol 2021; 153:211677. [PMID: 33439243 PMCID: PMC7809880 DOI: 10.1085/jgp.202012731] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022] Open
Abstract
It has become increasingly apparent that the lipid composition of cell membranes affects the function of transmembrane proteins such as ion channels. Here, we leverage the structural and functional diversity of small viral K+ channels to systematically examine the impact of bilayer composition on the pore module of single K+ channels. In vitro–synthesized channels were reconstituted into phosphatidylcholine bilayers ± cholesterol or anionic phospholipids (aPLs). Single-channel recordings revealed that a saturating concentration of 30% cholesterol had only minor and protein-specific effects on unitary conductance and gating. This indicates that channels have effective strategies for avoiding structural impacts of hydrophobic mismatches between proteins and the surrounding bilayer. In all seven channels tested, aPLs augmented the unitary conductance, suggesting that this is a general effect of negatively charged phospholipids on channel function. For one channel, we determined an effective half-maximal concentration of 15% phosphatidylserine, a value within the physiological range of aPL concentrations. The different sensitivity of two channel proteins to aPLs could be explained by the presence/absence of cationic amino acids at the interface between the lipid headgroups and the transmembrane domains. aPLs also affected gating in some channels, indicating that conductance and gating are uncoupled phenomena and that the impact of aPLs on gating is protein specific. In two channels, the latter can be explained by the altered orientation of the pore-lining transmembrane helix that prevents flipping of a phenylalanine side chain into the ion permeation pathway for long channel closings. Experiments with asymmetrical bilayers showed that this effect is leaflet specific and most effective in the inner leaflet, in which aPLs are normally present in plasma membranes. The data underscore a general positive effect of aPLs on the conductance of K+ channels and a potential interaction of their negative headgroup with cationic amino acids in their vicinity.
Collapse
Affiliation(s)
| | - Kerri Kukovetz
- Membrane Biophysics, Technische Universität Darmstadt, Darmstadt, Germany
| | - Ulf-Peter Hansen
- Department of Structural Biology, Christian-Albrechts-Universität, Kiel, Germany
| | - Indra Schroeder
- Membrane Biophysics, Technische Universität Darmstadt, Darmstadt, Germany
| | - James L Van Etten
- Department of Plant Pathology and Nebraska Center for Virology, University of Nebraska Lincoln, Lincoln, NE
| | - Anna Moroni
- Department of Biosciences and Consiglio Nazionale delle Ricerche, Istituto di Biofisica Milano, Università degli Studi di Milano, Milano, Italy
| | - Gerhard Thiel
- Membrane Biophysics, Technische Universität Darmstadt, Darmstadt, Germany
| | - Oliver Rauh
- Membrane Biophysics, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
95
|
Stewart TA, Hughes K, Stevenson AJ, Marino N, Ju AL, Morehead M, Davis FM. Mammary mechanobiology - investigating roles for mechanically activated ion channels in lactation and involution. J Cell Sci 2021; 134:jcs248849. [PMID: 33262312 DOI: 10.1242/jcs.248849] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/06/2020] [Indexed: 01/14/2023] Open
Abstract
The ability of a mother to produce a nutritionally complete neonatal food source has provided a powerful evolutionary advantage to mammals. Milk production by mammary epithelial cells is adaptive, its release is exquisitely timed, and its own glandular stagnation with the permanent cessation of suckling triggers the cell death and tissue remodeling that enables female mammals to nurse successive progeny. Chemical and mechanical signals both play a role in this process. However, despite this duality of input, much remains unknown about the nature and function of mechanical forces in this organ. Here, we characterize the force landscape in the functionally mature gland and the capacity of luminal and basal cells to experience and exert force. We explore molecular instruments for force-sensing, in particular channel-mediated mechanotransduction, revealing increased expression of Piezo1 in mammary tissue in lactation and confirming functional expression in luminal cells. We also reveal, however, that lactation and involution proceed normally in mice with luminal-specific Piezo1 deletion. These findings support a multifaceted system of chemical and mechanical sensing in the mammary gland, and a protective redundancy that ensures continued lactational competence and offspring survival.
Collapse
Affiliation(s)
- Teneale A Stewart
- Mater Research Institute-The University of Queensland, Faculty of Medicine, Woolloongabba, Queensland, 4102, Australia
- Translational Research Institute, Woolloongabba, Queensland, 4102, Australia
| | - Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, Cambridge, CB3 0ES, UK
| | - Alexander J Stevenson
- Mater Research Institute-The University of Queensland, Faculty of Medicine, Woolloongabba, Queensland, 4102, Australia
- Translational Research Institute, Woolloongabba, Queensland, 4102, Australia
| | - Natascia Marino
- Department of Medicine, Indiana University School of Medicine, Indianapolis, 46202, USA
- Susan G. Komen Tissue Bank at Indiana University Simon Cancer Center, Indianapolis, 46202, USA
| | - Adler L Ju
- Translational Research Institute, Woolloongabba, Queensland, 4102, Australia
| | - Michael Morehead
- Lane Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, 26506, USA
| | - Felicity M Davis
- Mater Research Institute-The University of Queensland, Faculty of Medicine, Woolloongabba, Queensland, 4102, Australia
- Translational Research Institute, Woolloongabba, Queensland, 4102, Australia
| |
Collapse
|
96
|
Doktorova M, Symons JL, Levental I. Structural and functional consequences of reversible lipid asymmetry in living membranes. Nat Chem Biol 2020; 16:1321-1330. [PMID: 33199908 PMCID: PMC7747298 DOI: 10.1038/s41589-020-00688-0] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022]
Abstract
Maintenance of lipid asymmetry across the two leaflets of the plasma membrane (PM) bilayer is a ubiquitous feature of eukaryotic cells. Loss of this asymmetry has been widely associated with cell death. However, increasing evidence points to the physiological importance of non-apoptotic, transient changes in PM asymmetry. Such transient scrambling events are associated with a range of biological functions, including intercellular communication and intracellular signaling. Thus, regulation of interleaflet lipid distribution in the PM is a broadly important but underappreciated cellular process with key physiological and structural consequences. Here, we compile the mounting evidence revealing multifaceted, functional roles of PM asymmetry and transient loss thereof. We discuss the consequences of reversible asymmetry on PM structure, biophysical properties and interleaflet coupling. We argue that despite widespread recognition of broad aspects of membrane asymmetry, its importance in cell biology demands more in-depth investigation of its features, regulation, and physiological and pathological implications.
Collapse
Affiliation(s)
| | - Jessica L Symons
- University of Texas Health Science Center at Houston, Houston, TX, USA
| | | |
Collapse
|
97
|
Buyan A, Cox CD, Barnoud J, Li J, Chan HSM, Martinac B, Marrink SJ, Corry B. Piezo1 Forms Specific, Functionally Important Interactions with Phosphoinositides and Cholesterol. Biophys J 2020; 119:1683-1697. [PMID: 32949489 PMCID: PMC7642233 DOI: 10.1016/j.bpj.2020.07.043] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/06/2020] [Accepted: 07/15/2020] [Indexed: 10/25/2022] Open
Abstract
Touch, hearing, and blood pressure regulation require mechanically gated ion channels that convert mechanical stimuli into electrical currents. One such channel is Piezo1, which plays a key role in the transduction of mechanical stimuli in humans and is implicated in diseases, such as xerocytosis and lymphatic dysplasia. There is building evidence that suggests Piezo1 can be regulated by the membrane environment, with the activity of the channel determined by the local concentration of lipids, such as cholesterol and phosphoinositides. To better understand the interaction of Piezo1 with its environment, we conduct simulations of the protein in a complex mammalian bilayer containing more than 60 different lipid types together with electrophysiology and mutagenesis experiments. We find that the protein alters its local membrane composition, enriching specific lipids and forming essential binding sites for phosphoinositides and cholesterol that are functionally relevant and often related to Piezo1-mediated pathologies. We also identify a number of key structural connections between the propeller and pore domains located close to lipid-binding sites.
Collapse
Affiliation(s)
- Amanda Buyan
- Research School of Biology, Australian National University, Acton, Canberra, Australia
| | - Charles D Cox
- Victor Chang Cardiac Research Institute, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia.
| | - Jonathan Barnoud
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Jinyuan Li
- Victor Chang Cardiac Research Institute, New South Wales, Australia
| | - Hannah S M Chan
- Research School of Biology, Australian National University, Acton, Canberra, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Siewert J Marrink
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Ben Corry
- Research School of Biology, Australian National University, Acton, Canberra, Australia.
| |
Collapse
|
98
|
Cox CD, Bavi N, Martinac B. Biophysical Principles of Ion-Channel-Mediated Mechanosensory Transduction. Cell Rep 2020; 29:1-12. [PMID: 31577940 DOI: 10.1016/j.celrep.2019.08.075] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/09/2019] [Accepted: 08/22/2019] [Indexed: 01/12/2023] Open
Abstract
Recent rapid progress in the field of mechanobiology has been driven by novel emerging tools and methodologies and growing interest from different scientific disciplines. Specific progress has been made toward understanding how cell mechanics is linked to intracellular signaling and the regulation of gene expression in response to a variety of mechanical stimuli. There is a direct link between the mechanoreceptors at the cell surface and intracellular biochemical signaling, which in turn controls downstream effector molecules. Among the mechanoreceptors in the cell membrane, mechanosensitive (MS) ion channels are essential for the ultra-rapid (millisecond) transduction of mechanical stimuli into biologically relevant signals. The three decades of research on mechanosensitive channels resulted in the formulation of two basic principles of mechanosensitive channel gating: force-from-lipids and force-from-filament. In this review, we revisit the biophysical principles that underlie the innate force-sensing ability of mechanosensitive channels as contributors to the force-dependent evolution of life forms.
Collapse
Affiliation(s)
- Charles D Cox
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW 2010, Australia
| | - Navid Bavi
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW 2010, Australia.
| |
Collapse
|
99
|
Pollet H, Cloos AS, Stommen A, Vanderroost J, Conrard L, Paquot A, Ghodsi M, Carquin M, Léonard C, Guthmann M, Lingurski M, Vermylen C, Killian T, Gatto L, Rider M, Pyr dit Ruys S, Vertommen D, Vikkula M, Brouillard P, Van Der Smissen P, Muccioli GG, Tyteca D. Aberrant Membrane Composition and Biophysical Properties Impair Erythrocyte Morphology and Functionality in Elliptocytosis. Biomolecules 2020; 10:biom10081120. [PMID: 32751168 PMCID: PMC7465299 DOI: 10.3390/biom10081120] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
Red blood cell (RBC) deformability is altered in inherited RBC disorders but the mechanism behind this is poorly understood. Here, we explored the molecular, biophysical, morphological, and functional consequences of α-spectrin mutations in a patient with hereditary elliptocytosis (pEl) almost exclusively expressing the Pro260 variant of SPTA1 and her mother (pElm), heterozygous for this mutation. At the molecular level, the pEI RBC proteome was globally preserved but spectrin density at cell edges was increased. Decreased phosphatidylserine vs. increased lysophosphatidylserine species, and enhanced lipid peroxidation, methemoglobin, and plasma acid sphingomyelinase (aSMase) activity were observed. At the biophysical level, although membrane transversal asymmetry was preserved, curvature at RBC edges and rigidity were increased. Lipid domains were altered for membrane:cytoskeleton anchorage, cholesterol content and response to Ca2+ exchange stimulation. At the morphological and functional levels, pEl RBCs exhibited reduced size and circularity, increased fragility and impaired membrane Ca2+ exchanges. The contribution of increased membrane curvature to the pEl phenotype was shown by mechanistic experiments in healthy RBCs upon lysophosphatidylserine membrane insertion. The role of lipid domain defects was proved by cholesterol depletion and aSMase inhibition in pEl. The data indicate that aberrant membrane content and biophysical properties alter pEl RBC morphology and functionality.
Collapse
Affiliation(s)
- Hélène Pollet
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Anne-Sophie Cloos
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Amaury Stommen
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Juliette Vanderroost
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Louise Conrard
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, 1200 Brussels, Belgium; (A.P.); (G.G.M.)
| | - Marine Ghodsi
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Mélanie Carquin
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Catherine Léonard
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Manuel Guthmann
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Maxime Lingurski
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Christiane Vermylen
- PEDI Unit, Institut de Recherche Expérimentale et Clinique & Saint-Luc Hospital, UCLouvain, 1200 Brussels, Belgium;
| | - Theodore Killian
- Computational Biology and Bioinformatics Unit, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (T.K.); (L.G.)
| | - Laurent Gatto
- Computational Biology and Bioinformatics Unit, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (T.K.); (L.G.)
| | - Mark Rider
- PHOS Unit & MASSPROT Proteomics Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (M.R.); (S.P.d.R.); (D.V.)
| | - Sébastien Pyr dit Ruys
- PHOS Unit & MASSPROT Proteomics Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (M.R.); (S.P.d.R.); (D.V.)
| | - Didier Vertommen
- PHOS Unit & MASSPROT Proteomics Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (M.R.); (S.P.d.R.); (D.V.)
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (M.V.); (P.B.)
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), de Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Pascal Brouillard
- Human Molecular Genetics, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (M.V.); (P.B.)
| | - Patrick Van Der Smissen
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, 1200 Brussels, Belgium; (A.P.); (G.G.M.)
| | - Donatienne Tyteca
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, 1200 Brussels, Belgium; (H.P.); (A.-S.C.); (A.S.); (J.V.); (L.C.); (M.G.); (M.C.); (C.L.); (M.G.); (M.L.); (P.V.D.S.)
- Correspondence:
| |
Collapse
|
100
|
Chang W, Fa H, Xiao D, Wang J. Targeting phosphatidylserine for Cancer therapy: prospects and challenges. Theranostics 2020; 10:9214-9229. [PMID: 32802188 PMCID: PMC7415799 DOI: 10.7150/thno.45125] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is a leading cause of mortality and morbidity worldwide. Despite major improvements in current therapeutic methods, ideal therapeutic strategies for improved tumor elimination are still lacking. Recently, immunotherapy has attracted much attention, and many immune-active agents have been approved for clinical use alone or in combination with other cancer drugs. However, some patients have a poor response to these agents. New agents and strategies are needed to overcome such deficiencies. Phosphatidylserine (PS) is an essential component of bilayer cell membranes and is normally present in the inner leaflet. In the physiological state, PS exposure on the external leaflet not only acts as an engulfment signal for phagocytosis in apoptotic cells but also participates in blood coagulation, myoblast fusion and immune regulation in nonapoptotic cells. In the tumor microenvironment, PS exposure is significantly increased on the surface of tumor cells or tumor cell-derived microvesicles, which have innate immunosuppressive properties and facilitate tumor growth and metastasis. To date, agents targeting PS have been developed, some of which are under investigation in clinical trials as combination drugs for various cancers. However, controversial results are emerging in laboratory research as well as in clinical trials, and the efficiency of PS-targeting agents remains uncertain. In this review, we summarize recent progress in our understanding of the physiological and pathological roles of PS, with a focus on immune suppressive features. In addition, we discuss current drug developments that are based on PS-targeting strategies in both experimental and clinical studies. We hope to provide a future research direction for the development of new agents for cancer therapy.
Collapse
Affiliation(s)
- Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
| | - Hongge Fa
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| | - Dandan Xiao
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| | - Jianxun Wang
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| |
Collapse
|