101
|
Blockade of integrin β3 signals to reverse the stem-like phenotype and drug resistance in melanoma. Cancer Chemother Pharmacol 2019; 83:615-624. [DOI: 10.1007/s00280-018-3760-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/24/2018] [Indexed: 01/06/2023]
|
102
|
Jetta D, Gottlieb PA, Verma D, Sachs F, Hua SZ. Shear stress induced nuclear shrinkage through activation of Piezo1 channels in epithelial cells. J Cell Sci 2019; 132:jcs.226076. [DOI: 10.1242/jcs.226076] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 04/29/2019] [Indexed: 12/30/2022] Open
Abstract
The cell nucleus responds to mechanical cues with changes in size, morphology, and motility. Previous work showed that external forces couple to nuclei through the cytoskeleton network, but we show here that changes in nuclear shape can be driven solely by calcium levels. Fluid shear stress applied to MDCK cells caused the nuclei to shrink through a Ca2+ dependent signaling pathway. Inhibiting mechanosensitive Piezo1 channels with GsMTx4 prevented nuclear shrinkage. Piezo1 knockdown also significantly reduced the nuclear shrinkage. Activation of Piezo1 with the agonist Yoda1 caused similar nucleus shrinkage without shear stress. These results demonstrate that Piezo1 channel is a key element for transmitting shear force input to nuclei. To ascertain the relative contributions of Ca2+ to cytoskeleton perturbation, we examined the F-actin reorganization under shear stress and static conditions, and showed that reorganization of the cytoskeleton is not necessary for nuclear shrinkage. These results emphasize the role of the mechanosensitive channels as primary transducers in force transmission to the nucleus.
Collapse
Affiliation(s)
- Deekshitha Jetta
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, New York 14260, USA
| | - Philip A. Gottlieb
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, New York 14260, USA
| | - Deepika Verma
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, New York 14260, USA
| | - Frederick Sachs
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, New York 14260, USA
| | - Susan Z. Hua
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, New York 14260, USA
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, New York 14260, USA
| |
Collapse
|
103
|
Velasco-Estevez M, Mampay M, Boutin H, Chaney A, Warn P, Sharp A, Burgess E, Moeendarbary E, Dev KK, Sheridan GK. Infection Augments Expression of Mechanosensing Piezo1 Channels in Amyloid Plaque-Reactive Astrocytes. Front Aging Neurosci 2018; 10:332. [PMID: 30405400 PMCID: PMC6204357 DOI: 10.3389/fnagi.2018.00332] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/01/2018] [Indexed: 01/07/2023] Open
Abstract
A defining pathophysiological hallmark of Alzheimer's disease (AD) is the amyloid plaque; an extracellular deposit of aggregated fibrillar Aβ1-42 peptides. Amyloid plaques are hard, brittle structures scattered throughout the hippocampus and cerebral cortex and are thought to cause hyperphosphorylation of tau, neurofibrillary tangles, and progressive neurodegeneration. Reactive astrocytes and microglia envelop the exterior of amyloid plaques and infiltrate their inner core. Glia are highly mechanosensitive cells and can almost certainly sense the mismatch between the normally soft mechanical environment of the brain and very stiff amyloid plaques via mechanosensing ion channels. Piezo1, a non-selective cation channel, can translate extracellular mechanical forces to intracellular molecular signaling cascades through a process known as mechanotransduction. Here, we utilized an aging transgenic rat model of AD (TgF344-AD) to study expression of mechanosensing Piezo1 ion channels in amyloid plaque-reactive astrocytes. We found that Piezo1 is upregulated with age in the hippocampus and cortex of 18-month old wild-type rats. However, more striking increases in Piezo1 were measured in the hippocampus of TgF344-AD rats compared to age-matched wild-type controls. Interestingly, repeated urinary tract infections with Escherichia coli bacteria, a common comorbidity in elderly people with dementia, caused further elevations in Piezo1 channel expression in the hippocampus and cortex of TgF344-AD rats. Taken together, we report that aging and peripheral infection augment amyloid plaque-induced upregulation of mechanoresponsive ion channels, such as Piezo1, in astrocytes. Further research is required to investigate the role of astrocytic Piezo1 in the Alzheimer's brain, whether modulating channel opening will protect or exacerbate the disease state, and most importantly, if Piezo1 could prove to be a novel drug target for age-related dementia.
Collapse
Affiliation(s)
- María Velasco-Estevez
- Neuroimmulology & Neurotherapeutics Laboratory, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
- Drug Development, Department of Physiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Myrthe Mampay
- Neuroimmulology & Neurotherapeutics Laboratory, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Hervé Boutin
- Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
| | - Aisling Chaney
- Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
- Department of Radiology, Stanford University, Stanford, CA, United States
| | - Peter Warn
- Evotec (UK) Ltd., Manchester Science Park, Manchester, United Kingdom
| | - Andrew Sharp
- Evotec (UK) Ltd., Manchester Science Park, Manchester, United Kingdom
| | - Ellie Burgess
- Evotec (UK) Ltd., Manchester Science Park, Manchester, United Kingdom
| | - Emad Moeendarbary
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Kumlesh K. Dev
- Drug Development, Department of Physiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Graham K. Sheridan
- Neuroimmulology & Neurotherapeutics Laboratory, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| |
Collapse
|
104
|
Piezo proteins: incidence and abundance in the enteric nervous system. Is there a link with mechanosensitivity? Cell Tissue Res 2018; 375:605-618. [PMID: 30324494 DOI: 10.1007/s00441-018-2926-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/10/2018] [Indexed: 10/28/2022]
Abstract
Piezo channels play fundamental roles in many physiological processes. Their presence and functional role in the enteric nervous system is still not known. We hypothesize that they play a role in mechanotransduction in enteric neurons. Our aims are to quantify the presence of both Piezo1 and 2 in enteric neurons throughout the gastrointestinal tract using immunohistochemistry and analyze their function(s) using neuroimaging techniques and pharmacological investigations. In order to perform a systematic and comparative study, we performed our experiments in gastrointestinal tissue from guinea pigs, mice and humans. Piezo1 (20-70%) is expressed by both enteric neuronal cell bodies and fibers in the myenteric and submucosal plexi of all the species investigated. Generally, Piezo1 expressing somata are more numerous in the submucosal plexus (50-80%) than in the myenteric plexus (15-35%) apart from the stomach where Piezo1 is expressed in up to 60% of cell bodies. Myenteric Piezo1 neurons mainly (60-100%) but not exclusively, also express nitric oxide synthase, a minority express choline acetyltransferase. In the submucosal plexus, Piezo1 neurons co-express vasoactive intestinal peptide (40-90%). Conversely, expression of Piezo2 is extremely rare in the somata of enteric neurons and is present in few neurites. In functional experiments, 38-76% of the mechanosensitive neurons expressed Piezo1 channels. Statistical analysis showed a positive significant correlation between mechanosensitive and Piezo1 positive neurons. However, pharmacological experiments using an activator and an inhibitor of Piezo channels did not demonstrate changes in mechanotransduction. A major role of Piezo1 in the mechanosensitivity of enteric neurons can be excluded.
Collapse
|
105
|
Maneshi MM, Ziegler L, Sachs F, Hua SZ, Gottlieb PA. Enantiomeric Aβ peptides inhibit the fluid shear stress response of PIEZO1. Sci Rep 2018; 8:14267. [PMID: 30250223 PMCID: PMC6155315 DOI: 10.1038/s41598-018-32572-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 09/06/2018] [Indexed: 01/23/2023] Open
Abstract
Traumatic brain injury (TBI) elevates Abeta (Aβ) peptides in the brain and cerebral spinal fluid. Aβ peptides are amphipathic molecules that can modulate membrane mechanics. Because the mechanosensitive cation channel PIEZO1 is gated by membrane tension and curvature, it prompted us to test the effects of Aβ on PIEZO1. Using precision fluid shear stress as a stimulus, we found that Aβ monomers inhibit PIEZO1 at femtomolar to picomolar concentrations. The Aβ oligomers proved much less potent. The effect of Aβs on Piezo gating did not involve peptide-protein interactions since the D and L enantiomers had similar effects. Incubating a fluorescent derivative of Aβ and a fluorescently tagged PIEZO1, we showed that Aβ can colocalize with PIEZO1, suggesting that they both had an affinity for particular regions of the bilayer. To better understand the PIEZO1 inhibitory effects of Aβ, we examined their effect on wound healing. We observed that over-expression of PIEZO1 in HEK293 cells increased cell migration velocity ~10-fold, and both enantiomeric Aβ peptides and GsMTx4 independently inhibited migration, demonstrating involvement of PIEZO1 in cell motility. As part of the motility study we examined the correlation of PIEZO1 function with tension in the cytoskeleton using a genetically encoded fluorescent stress probe. Aβ peptides increased resting stress in F-actin, and is correlated with Aβ block of PIEZO1-mediated Ca2+ influx. Aβ inhibition of PIEZO1 in the absence of stereospecific peptide-protein interactions shows that Aβ peptides modulate both cell membrane and cytoskeletal mechanics to control PIEZO1-triggered Ca2+ influx.
Collapse
Affiliation(s)
- Mohammad M Maneshi
- Department of Physiology and Biophysics, 302 Cary Hall, State University of New York at Buffalo, Buffalo, NY, 14214, USA
- Department of Mechanical and Aerospace Engineering, 340 Jarvis Hall, State University of New York at Buffalo, Buffalo, New York, 14260, USA
- 745 N Fairbanks, Tarry 7-718, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Lynn Ziegler
- Department of Physiology and Biophysics, 302 Cary Hall, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Frederick Sachs
- Department of Physiology and Biophysics, 302 Cary Hall, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Susan Z Hua
- Department of Physiology and Biophysics, 302 Cary Hall, State University of New York at Buffalo, Buffalo, NY, 14214, USA
- Department of Mechanical and Aerospace Engineering, 340 Jarvis Hall, State University of New York at Buffalo, Buffalo, New York, 14260, USA
| | - Philip A Gottlieb
- Department of Physiology and Biophysics, 302 Cary Hall, State University of New York at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
106
|
Zhang J, Zhou Y, Huang T, Wu F, Liu L, Kwan JSH, Cheng ASL, Yu J, To KF, Kang W. PIEZO1 functions as a potential oncogene by promoting cell proliferation and migration in gastric carcinogenesis. Mol Carcinog 2018; 57:1144-1155. [DOI: 10.1002/mc.22831] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Jinglin Zhang
- Department of Anatomical and Cellular Pathology; State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong; Hong Kong SAR PR China
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Li Ka Shing Institute of Health Science; Sir Y.K. Pao Cancer Center; The Chinese University of Hong Kong; Hong Kong SAR PR China
| | - Yuhang Zhou
- Department of Anatomical and Cellular Pathology; State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong; Hong Kong SAR PR China
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Li Ka Shing Institute of Health Science; Sir Y.K. Pao Cancer Center; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen PR China
| | - Tingting Huang
- Department of Anatomical and Cellular Pathology; State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong; Hong Kong SAR PR China
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Li Ka Shing Institute of Health Science; Sir Y.K. Pao Cancer Center; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen PR China
| | - Feng Wu
- Department of Anatomical and Cellular Pathology; State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong; Hong Kong SAR PR China
| | - Liping Liu
- Department of Hepatobiliary and Pancreatic Surgery; Shenzhen People's Hospital; Second Clinical Medical College of Jinan University; Shenzhen Guangdong Province PR China
| | - Johnny S. H. Kwan
- Department of Anatomical and Cellular Pathology; State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong; Hong Kong SAR PR China
| | - Alfred S. L. Cheng
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen PR China
- School of Biomedical Sciences; The Chinese University of Hong Kong; Hong Kong PR China
| | - Jun Yu
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen PR China
- Department of Medicine and Therapeutics; The Chinese University of Hong Kong; Hong Kong PR China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology; State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong; Hong Kong SAR PR China
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Li Ka Shing Institute of Health Science; Sir Y.K. Pao Cancer Center; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen PR China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology; State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong; Hong Kong SAR PR China
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Li Ka Shing Institute of Health Science; Sir Y.K. Pao Cancer Center; The Chinese University of Hong Kong; Hong Kong SAR PR China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen PR China
| |
Collapse
|
107
|
PIEZO1 Channel Is a Potential Regulator of Synovial Sarcoma Cell-Viability. Int J Mol Sci 2018; 19:ijms19051452. [PMID: 29757938 PMCID: PMC5983681 DOI: 10.3390/ijms19051452] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/03/2018] [Accepted: 05/10/2018] [Indexed: 01/02/2023] Open
Abstract
Detection of mechanical stress is essential for diverse biological functions including touch, audition, and maintenance of vascular myogenic tone. PIEZO1, a mechano-sensing cation channel, is widely expressed in neuronal and non-neuronal cells and is expected to be involved in important biological functions. Here, we examined the possibility that PIEZO1 is involved in the regulation of synovial sarcoma cell-viability. Application of a PIEZO1 agonist Yoda1 effectively induced Ca2+ response and cation channel currents in PIEZO1-expressing HEK (HEK-Piezo1) cells and synovial sarcoma SW982 (SW982) cells. Mechanical stress, as well as Yoda1, induced the activity of an identical channel of conductance with 21.6 pS in HEK-Piezo1 cells. In contrast, Yoda1 up to 10 μM had no effects on membrane currents in HEK cells without transfecting PIEZO1. A knockdown of PIEZO1 with siRNA in SW982 cells abolished Yoda1-induced Ca2+ response and significantly reduced cell cell-viability. Because PIEZO1 is highly expressed in SW982 cells and its knockdown affects cell-viability, this gene is a potential target against synovial sarcoma.
Collapse
|
108
|
Narayanan P, Hütte M, Kudryasheva G, Taberner FJ, Lechner SG, Rehfeldt F, Gomez-Varela D, Schmidt M. Myotubularin related protein-2 and its phospholipid substrate PIP 2 control Piezo2-mediated mechanotransduction in peripheral sensory neurons. eLife 2018. [PMID: 29521261 PMCID: PMC5898911 DOI: 10.7554/elife.32346] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Piezo2 ion channels are critical determinants of the sense of light touch in vertebrates. Yet, their regulation is only incompletely understood. We recently identified myotubularin related protein-2 (Mtmr2), a phosphoinositide (PI) phosphatase, in the native Piezo2 interactome of murine dorsal root ganglia (DRG). Here, we demonstrate that Mtmr2 attenuates Piezo2-mediated rapidly adapting mechanically activated (RA-MA) currents. Interestingly, heterologous Piezo1 and other known MA current subtypes in DRG appeared largely unaffected by Mtmr2. Experiments with catalytically inactive Mtmr2, pharmacological blockers of PI(3,5)P2 synthesis, and osmotic stress suggest that Mtmr2-dependent Piezo2 inhibition involves depletion of PI(3,5)P2. Further, we identified a PI(3,5)P2 binding region in Piezo2, but not Piezo1, that confers sensitivity to Mtmr2 as indicated by functional analysis of a domain-swapped Piezo2 mutant. Altogether, our results propose local PI(3,5)P2 modulation via Mtmr2 in the vicinity of Piezo2 as a novel mechanism to dynamically control Piezo2-dependent mechanotransduction in peripheral sensory neurons. We often take our sense of touch for granted. Yet, our every-day life greatly depends on the ability to perceive our environment to alert us of danger or to further social interactions, such as mother-child bonding. Our sense of touch relies on the conversion of mechanical stimuli to electrical signals (this is known as mechanotransduction), which then travel to brain to be processed. This task is fulfilled by specific ion channels called Piezo2, which are activated when cells are exposed to pressure and other mechanical forces. These channels can be found in sensory nerves and specialized structures in the skin, where they help to detect physical contact, roughness of surfaces and the position of our body parts. It is still not clear how Piezo2 channels are regulated but previous research by several laboratories suggests that they work in conjunction with other proteins. One of these proteins is the myotubularin related protein-2, or Mtmr2 for short. Now, Narayanan et al. – including some of the researchers involved in the previous research – set out to advance our understanding of the molecular basis of touch and looked more closely at Mtmr2. To test if Mtmr2 played a role in mechanotransduction, Narayanan et al. both increased and reduced the levels of this protein in sensory neurons of mice grown in the laboratory. When Mtmr2 levels were low, the activity of Piezo2 channels increased. However, when the protein levels were high, Piezo2 channels were inhibited. These results suggest that Mtmr2 can control the activity of Piezo2. Further experiments, in which Mtmr2 was genetically modified or sensory neurons were treated with chemicals, revealed that Mtmr2 reduces a specific fatty acid in the membrane of nerve cells, which in turn attenuates the activity of Piezo2. This study identified Mtmr2 and distinct fatty acids in the cell membrane as new components of the complex setup required for the sense of touch. A next step will be to test if these molecules also influence the activity of Piezo2 when the skin has become injured or upon inflammation.
Collapse
Affiliation(s)
- Pratibha Narayanan
- Emmy Noether-Group Somatosensory Signaling and Systems Biology, Max Planck Institute for Experimental Medicine, Goettingen, Germany
| | - Meike Hütte
- Emmy Noether-Group Somatosensory Signaling and Systems Biology, Max Planck Institute for Experimental Medicine, Goettingen, Germany
| | - Galina Kudryasheva
- Third Institute of Physics - Biophysics, University of Goettingen, Goettingen, Germany
| | | | | | - Florian Rehfeldt
- Third Institute of Physics - Biophysics, University of Goettingen, Goettingen, Germany
| | - David Gomez-Varela
- Emmy Noether-Group Somatosensory Signaling and Systems Biology, Max Planck Institute for Experimental Medicine, Goettingen, Germany
| | - Manuela Schmidt
- Emmy Noether-Group Somatosensory Signaling and Systems Biology, Max Planck Institute for Experimental Medicine, Goettingen, Germany
| |
Collapse
|
109
|
Piezo2 channel regulates RhoA and actin cytoskeleton to promote cell mechanobiological responses. Proc Natl Acad Sci U S A 2018; 115:1925-1930. [PMID: 29432180 DOI: 10.1073/pnas.1718177115] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Actin polymerization and assembly into stress fibers (SFs) is central to many cellular processes. However, how SFs form in response to the mechanical interaction of cells with their environment is not fully understood. Here we have identified Piezo2 mechanosensitive cationic channel as a transducer of environmental physical cues into mechanobiological responses. Piezo2 is needed by brain metastatic cells from breast cancer (MDA-MB-231-BrM2) to probe their physical environment as they anchor and pull on their surroundings or when confronted with confined migration through narrow pores. Piezo2-mediated Ca2+ influx activates RhoA to control the formation and orientation of SFs and focal adhesions (FAs). A possible mechanism for the Piezo2-mediated activation of RhoA involves the recruitment of the Fyn kinase to the cell leading edge as well as calpain activation. Knockdown of Piezo2 in BrM2 cells alters SFs, FAs, and nuclear translocation of YAP; a phenotype rescued by overexpression of dominant-positive RhoA or its downstream effector, mDia1. Consequently, hallmarks of cancer invasion and metastasis related to RhoA, actin cytoskeleton, and/or force transmission, such as migration, extracellular matrix degradation, and Serpin B2 secretion, were reduced in cells lacking Piezo2.
Collapse
|
110
|
Maffioli E, Schulte C, Nonnis S, Grassi Scalvini F, Piazzoni C, Lenardi C, Negri A, Milani P, Tedeschi G. Proteomic Dissection of Nanotopography-Sensitive Mechanotransductive Signaling Hubs that Foster Neuronal Differentiation in PC12 Cells. Front Cell Neurosci 2018; 11:417. [PMID: 29354032 PMCID: PMC5758595 DOI: 10.3389/fncel.2017.00417] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
Neuronal cells are competent in precisely sensing nanotopographical features of their microenvironment. The perceived microenvironmental information will be “interpreted” by mechanotransductive processes and impacts on neuronal functioning and differentiation. Attempts to influence neuronal differentiation by engineering substrates that mimic appropriate extracellular matrix (ECM) topographies are hampered by the fact that profound details of mechanosensing/-transduction complexity remain elusive. Introducing omics methods into these biomaterial approaches has the potential to provide a deeper insight into the molecular processes and signaling cascades underlying mechanosensing/-transduction but their exigence in cellular material is often opposed by technical limitations of major substrate top-down fabrication methods. Supersonic cluster beam deposition (SCBD) allows instead the bottom-up fabrication of nanostructured substrates over large areas characterized by a quantitatively controllable ECM-like nanoroughness that has been recently shown to foster neuron differentiation and maturation. Exploiting this capacity of SCBD, we challenged mechanosensing/-transduction and differentiative behavior of neuron-like PC12 cells with diverse nanotopographies and/or changes of their biomechanical status, and analyzed their phosphoproteomic profiles in these settings. Versatile proteins that can be associated to significant processes along the mechanotransductive signal sequence, i.e., cell/cell interaction, glycocalyx and ECM, membrane/f-actin linkage and integrin activation, cell/substrate interaction, integrin adhesion complex, actomyosin organization/cellular mechanics, nuclear organization, and transcriptional regulation, were affected. The phosphoproteomic data suggested furthermore an involvement of ILK, mTOR, Wnt, and calcium signaling in these nanotopography- and/or cell mechanics-related processes. Altogether, potential nanotopography-sensitive mechanotransductive signaling hubs participating in neuronal differentiation were dissected.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Carsten Schulte
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Simona Nonnis
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Claudio Piazzoni
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Cristina Lenardi
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Armando Negri
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Paolo Milani
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| |
Collapse
|
111
|
Minchin JEN, Rawls JF. Elucidating the role of plexin D1 in body fat distribution and susceptibility to metabolic disease using a zebrafish model system. Adipocyte 2017; 6:277-283. [PMID: 28792859 DOI: 10.1080/21623945.2017.1356504] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Non-communicable diseases (NCDs) such as cardiovascular disease, diabetes and cancer were responsible for 68% of all deaths worldwide in 2012. The regional distribution of lipid deposited within adipose tissue (AT) - so called body fat distribution (BFD) - is a strong risk factor for NCDs. BFD is highly heritable; however, the genetic basis of BFD is almost entirely unknown. Genome-wide association studies have identified several loci associated with BFD, including at Plexin D1 (PLXND1) - a gene known to modulate angiogenesis. We recently demonstrated that zebrafish homozygous for a null mutation in plxnd1 had a reduced capacity to store lipid in visceral AT (VAT) leading to altered BFD. Moreover, we found that type V collagens were upregulated in plxnd1 mutants, and mediated the inhibitory effect of Plxnd1 on VAT growth. These results strengthen evidence that Plxnd1 influences BFD in human populations, and validate zebrafish as a model to study BFD. However, many pertinent questions remain unanswered. Here we outline potential Plxnd1 mechanisms of action in AT, and describe the genetic architecture at human PLXND1 that is associated with BFD and NCD susceptibility.
Collapse
Affiliation(s)
- James E. N. Minchin
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| |
Collapse
|
112
|
A thirty-year quest for a role of R-Ras in cancer: from an oncogene to a multitasking GTPase. Cancer Lett 2017; 403:59-65. [DOI: 10.1016/j.canlet.2017.06.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/28/2017] [Accepted: 06/03/2017] [Indexed: 12/30/2022]
|
113
|
Zhao Q, Wu K, Chi S, Geng J, Xiao B. Heterologous Expression of the Piezo1-ASIC1 Chimera Induces Mechanosensitive Currents with Properties Distinct from Piezo1. Neuron 2017; 94:274-277. [PMID: 28426963 DOI: 10.1016/j.neuron.2017.03.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/22/2017] [Accepted: 03/27/2017] [Indexed: 01/30/2023]
Abstract
Piezo1 represents a prototype of the mammalian mechanosensitive cation channel, but its molecular mechanism remains elusive. In a recent study, we showed that C-terminal region, which contains the last two TMs, of 2189-2547 of Piezo1 forms the bona fide pore module, and systematically identified the pore-lining helix and key pore-property-determining residues (Zhao et al., 2016). Furthermore, we have engineered the Piezo1(1-2190)-ASIC1 chimera (fusing the N-terminal region of 1-2190 to the mechano-insensitive ASIC1) that mediated mechanical- and acid-evoked currents in HEK293T cells, indicating the sufficiency of the N-terminal region in mechanotransduction. Now in a Matters Arising, the authors specifically questioned the implication of the chimera data among the many findings shown in our paper. They replicated the chimera-mediated mechanosensitive currents in HEK293T cells that have nearly no detectable expression of endogenous Piezo1, but paradoxically found the chimera to be less effective in Piezo1 knockout HEK293T cells, indicating the involvement of endogenous Piezo1. In this Matters Arising Response, we discuss the chimera results and consider potential interpretations in light of the Matters Arising from Dubin et al. (2017), published concurrently in this issue of Neuron. Please see also the response from Hong et al. (2017), published in this issue.
Collapse
Affiliation(s)
- Qiancheng Zhao
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Kun Wu
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Shaopeng Chi
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Jie Geng
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Bailong Xiao
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
114
|
Nourse JL, Pathak MM. How cells channel their stress: Interplay between Piezo1 and the cytoskeleton. Semin Cell Dev Biol 2017; 71:3-12. [PMID: 28676421 DOI: 10.1016/j.semcdb.2017.06.018] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/23/2017] [Accepted: 06/25/2017] [Indexed: 11/17/2022]
Abstract
Cells constantly encounter mechanical stimuli in their environment, such as dynamic forces and mechanical features of the extracellular matrix. These mechanical cues are transduced into biochemical signals, and integrated with genetic and chemical signals to modulate diverse physiological processes. Cells also actively generate forces to internally transport cargo, to explore the physical properties of their environment and to spatially position themselves and other cells during development. Mechanical forces are therefore central to development, homeostasis, and repair. Several molecular and biophysical strategies are utilized by cells for detecting and generating mechanical forces. Here we discuss an important class of molecules involved in sensing and transducing mechanical forces - mechanically-activated ion channels. We focus primarily on the Piezo1 ion channel, and examine its relationship with the cellular cytoskeleton.
Collapse
Affiliation(s)
- Jamison L Nourse
- Department of Physiology & Biophysics, Sue & Bill Gross Stem Cell Research Center, 835 Health Sciences Road, Room 275B, UC Irvine, Irvine, CA 92697, United States
| | - Medha M Pathak
- Department of Physiology & Biophysics, Sue & Bill Gross Stem Cell Research Center, 835 Health Sciences Road, Room 275B, UC Irvine, Irvine, CA 92697, United States.
| |
Collapse
|
115
|
Scholz N, Monk KR, Kittel RJ, Langenhan T. Adhesion GPCRs as a Putative Class of Metabotropic Mechanosensors. Handb Exp Pharmacol 2017; 234:221-247. [PMID: 27832490 DOI: 10.1007/978-3-319-41523-9_10] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Adhesion GPCRs as mechanosensors. Different aGPCR homologs and their cognate ligands have been described in settings, which suggest that they function in a mechanosensory capacity. For details, see text G protein-coupled receptors (GPCRs) constitute the most versatile superfamily of biosensors. This group of receptors is formed by hundreds of GPCRs, each of which is tuned to the perception of a specific set of stimuli a cell may encounter emanating from the outside world or from internal sources. Most GPCRs are receptive for chemical compounds such as peptides, proteins, lipids, nucleotides, sugars, and other organic compounds, and this capacity is utilized in several sensory organs to initiate visual, olfactory, gustatory, or endocrine signals. In contrast, GPCRs have only anecdotally been implicated in the perception of mechanical stimuli. Recent studies, however, show that the family of adhesion GPCRs (aGPCRs), which represents a large panel of over 30 homologs within the GPCR superfamily, displays molecular design and expression patterns that are compatible with receptivity toward mechanical cues (Fig. 1). Here, we review physiological and molecular principles of established mechanosensors, discuss their relevance for current research of the mechanosensory function of aGPCRs, and survey the current state of knowledge on aGPCRs as mechanosensing molecules.
Collapse
Affiliation(s)
- Nicole Scholz
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Röntgenring 9, Würzburg, 97070, Germany.
| | - Kelly R Monk
- Department of Developmental Biology, Hope Center for Neurologic Disorders, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Robert J Kittel
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Röntgenring 9, Würzburg, 97070, Germany
| | - Tobias Langenhan
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Röntgenring 9, Würzburg, 97070, Germany.
| |
Collapse
|
116
|
Abstract
Mutations in the genes encoding the mechanosensitive cation channels PIEZO1 and PIEZO2 are responsible for multiple hereditary human diseases. Loss-of-function mutations in the human PIEZO1 gene cause autosomal recessive congenital lymphatic dysplasia. Gain-of-function mutations in the human PIEZO1 gene cause the autosomal dominant hemolytic anemia, hereditary xerocytosis (also known as dehydrated stomatocytosis). Loss-of-function mutations in the human PIEZO2 gene cause an autosomal recessive syndrome of muscular atrophy with perinatal respiratory distress, arthrogryposis, and scoliosis. Gain-of-function mutations in the human PIEZO2 gene cause three clinical types of autosomal dominant distal arthrogryposis. This chapter will review the hereditary diseases caused by mutations in the PIEZO genes and will discuss additional physiological systems in which PIEZO channel dysfunction may contribute to human disease pathophysiology.
Collapse
Affiliation(s)
- S L Alper
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
117
|
Gnanasambandam R, Gottlieb PA, Sachs F. The Kinetics and the Permeation Properties of Piezo Channels. CURRENT TOPICS IN MEMBRANES 2017; 79:275-307. [PMID: 28728821 DOI: 10.1016/bs.ctm.2016.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Piezo channels are eukaryotic, cation-selective mechanosensitive channels (MSCs), which show rapid activation and voltage-dependent inactivation. The kinetics of these channels are largely consistent across multiple cell types and different stimulation paradigms with some minor variability. No accessory subunits that associate with Piezo channels have been reported. They are homotrimers and each ∼300kD monomer has an N-terminal propeller blade-like mechanosensing module, which can confer mechanosensing capabilities on ASIC-1 (the trimeric non-MSC, acid-sensing ion channel-1) and a C-terminal pore module, which influences conductance, selectivity, and channel inactivation. Repeated stimulation can cause domain fracture and diffusion of these channels leading to synchronous loss of inactivation. The reconstituted channels spontaneously open only in asymmetric bilayers but lack inactivation. Mutations that cause hereditary xerocytosis alter PIEZO1 kinetics. The kinetics of the wild-type PIEZO1 and alterations thereof in mutants (M2225R, R2456K, and DhPIEZO1) are summarized in the form of a quantitative model and hosted online. The pore is permeable to alkali ions although Li+ permeates poorly. Divalent cations, notably Ca2+, traverse the channel and inhibit the flux of monovalents. The large monovalent organic cations such as tetramethyl ammonium and tetraethyl ammonium can traverse the channel, but slowly, suggesting a pore diameter of ∼8Å, and the estimated in-plane area change upon opening is around 6-20nm2. Ruthenium red can enter the channel only from the extracellular side and seems to bind in a pocket close to residue 2496.
Collapse
Affiliation(s)
- R Gnanasambandam
- State University of New York at Buffalo, Buffalo, NY, United States
| | - P A Gottlieb
- State University of New York at Buffalo, Buffalo, NY, United States
| | - F Sachs
- State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
118
|
Hyman AJ, Tumova S, Beech DJ. Piezo1 Channels in Vascular Development and the Sensing of Shear Stress. CURRENT TOPICS IN MEMBRANES 2017; 79:37-57. [PMID: 28728823 DOI: 10.1016/bs.ctm.2016.11.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A critical point in mammalian development occurs before mid-embryogenesis when the heart starts to beat, pushing blood into the nascent endothelial lattice. This pushing force is a signal, detected by endothelial cells as a frictional force (shear stress) to trigger cellular changes that underlie the essential processes of vascular remodeling and expansion required for embryonic growth. The processes are complex and multifactorial and Piezo1 became a recognized player only 2years ago, 4years after Piezo1's initial discovery as a functional membrane protein. Piezo1 is now known to be critical in murine embryonic development just at the time when the pushing force is first detected by endothelial cells. Murine Piezo1 gene disruption in endothelial cells is embryonic lethal and mutations in human PIEZO1 associate with severe disease phenotype due to abnormal lymphatic vascular development. Piezo1 proteins coassemble to form calcium-permeable nonselective cationic channels, most likely as trimers. They are large proteins with little if any resemblance to other proteins or ion channel subunits. The channels appear to sense mechanical force directly, including the force imposed on endothelial cells by physiological shear stress. Here, we review current knowledge of Piezo1 in the vascular setting and discuss hypotheses about how it might serve its vascular functions and integrate with other mechanisms. Piezo1 is a new important player for investigators in this field and promises much as a basis for better understanding of vascular physiology and pathophysiology and perhaps also discovery of new therapies.
Collapse
Affiliation(s)
- A J Hyman
- University of Leeds, Leeds, United Kingdom
| | - S Tumova
- University of Leeds, Leeds, United Kingdom
| | - D J Beech
- University of Leeds, Leeds, United Kingdom
| |
Collapse
|
119
|
|
120
|
Abstract
Mechanotransduction is one of the processes by which cells sense and convert mechanical stimuli into biological signals. Experimental data from various species have revealed crucial roles for mechanotransduction in organ development and a plethora of physiological activities. Piezo proteins have recently been identified as the long-sought-after mechanically activated cation channels in eukaryotes. The architecture of mouse Piezo1 (mPiezo1) channel determined by cryoelectron microscopic single-particle analysis at medium resolution yielded important insights into the mechanical force sensing mechanism. mPiezo1 is found to form a trimeric propeller-like structure with the extracellular domains resembling three distal blades and a central cap. The transmembrane region consists of a central pore module that likely determines the ion-conducting properties of mPiezo1, and three peripheral wings formed by arrays of paired transmembrane helices. Compared with the central pore module, the three distal blades display considerably larger flexibility. In the intracellular region, three long beam-like domains (∼80Å in length) support the whole transmembrane region and connect the mobile peripheral regions to the central pore module. This unique design suggests that the trimeric mPiezo1 may mechanistically function in similar principles as how propellers sense and transduce force to control the ion conductivity. This review summarizes the current knowledge on the structure and proposes possible gating mechanisms of mPiezo1.
Collapse
|
121
|
Abstract
PIEZO1 is a mechanosensitive eukaryotic cation-selective channel that rapidly inactivates in a voltage-dependent manner. We previously showed that a fluorescent protein could be encoded within the hPIEZO1 sequence without loss of function. In this work, we split the channel into two at this site and asked if coexpression would produce a functional channel or whether gating and permeation might be contained in either segment. The split protein was expressed in two segments by a bicistronic plasmid where the first segment spanned residues 1 to 1591, and the second segment spanned 1592 to 2521. When the "split protein" is coexpressed, the parts associate to form a normal channel. We measured the whole-cell, cell-attached and outside-out patch currents in transfected HEK293 cells. Indentation produced whole-cell currents monotonic with the stimulus. Single channel recordings showed voltage-dependent inactivation. The Boltzmann activation curve for outside-out patches had a slope of 8.6/mmHg vs 8.1 for wild type, and a small leftward shift in the midpoint (32 mmHg vs 41 mmHg). The association of the two channel domains was confirmed by FRET measurements of mCherry on the N-terminus and EGFP on the C-terminus. Neither of the individual protein segments produced current when expressed alone.
Collapse
|
122
|
Yan X, Yan M, Guo Y, Singh G, Chen Y, Yu M, Wang D, Hillery CA, Chan AM. R-Ras Regulates Murine T Cell Migration and Intercellular Adhesion Molecule-1 Binding. PLoS One 2015; 10:e0145218. [PMID: 26710069 PMCID: PMC4692399 DOI: 10.1371/journal.pone.0145218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 11/30/2015] [Indexed: 12/04/2022] Open
Abstract
The trafficking of T-lymphocytes to peripheral draining lymph nodes is crucial for mounting an adaptive immune response. The role of chemokines in the activation of integrins via Ras-related small GTPases has been well established. R-Ras is a member of the Ras-subfamily of small guanosine-5’-triphosphate-binding proteins and its role in T cell trafficking has been investigated in R-Ras null mice (Rras−/−). An examination of the lymphoid organs of Rras−/− mice revealed a 40% reduction in the cellularity of the peripheral lymph nodes. Morphologically, the high endothelial venules of Rras−/− mice were more disorganized and less mature than those of wild-type mice. Furthermore, CD4+ and CD8+ T cells from Rras−/− mice had approximately 42% lower surface expression of L-selectin/CD62L. These aberrant peripheral lymph node phenotypes were associated with proliferative and trafficking defects in Rras−/− T cells. Furthermore, R-Ras could be activated by the chemokine, CCL21. Indeed, Rras−/− T cells had approximately 14.5% attenuation in binding to intercellular adhesion molecule 1 upon CCL21 stimulation. Finally, in a graft-versus host disease model, recipient mice that were transfused with Rras−/− T cells showed a significant reduction in disease severity when compared with mice transplanted with wild-type T cells. These findings implicate a role for R-Ras in T cell trafficking in the high endothelial venules during an effective immune response.
Collapse
Affiliation(s)
- Xiaocai Yan
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Mingfei Yan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Yihe Guo
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Gobind Singh
- Department of Oncological Sciences, The Mount Sinai School of Medicine, New York, New York, United States of America
| | - Yuhong Chen
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Mei Yu
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Demin Wang
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Cheryl A Hillery
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America.,Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Andrew M Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| |
Collapse
|
123
|
Dupont S. Role of YAP/TAZ in cell-matrix adhesion-mediated signalling and mechanotransduction. Exp Cell Res 2015; 343:42-53. [PMID: 26524510 DOI: 10.1016/j.yexcr.2015.10.034] [Citation(s) in RCA: 346] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 10/29/2015] [Indexed: 12/12/2022]
Abstract
Signalling from the extracellular matrix (ECM) is a fundamental cellular input that sustains proliferation, opposes cell death and regulates differentiation. Through integrins, cells perceive both the chemical composition and physical properties of the ECM. In particular, cell behaviour is profoundly influenced by the mechanical elasticity or stiffness of the ECM, which regulates the ability of cells to develop forces through their contractile actomyosin cytoskeleton and to mature focal adhesions. This mechanosensing ability affects fundamental cellular functions, such that alterations of ECM stiffness is nowadays considered not a simple consequence of pathology, but a causative input driving aberrant cell behaviours. We here discuss recent advances on how mechanical signals intersect nuclear transcription and in particular the activity of YAP/TAZ transcriptional coactivators, known downstream transducers of the Hippo pathway and important effectors of ECM mechanical cues.
Collapse
Affiliation(s)
- Sirio Dupont
- Department of Molecular Medicine, University of Padua Medical School, via Bassi 58/B, 35131 Padua, Italy.
| |
Collapse
|
124
|
Andolfo I, Russo R, Manna F, Shmukler BE, Gambale A, Vitiello G, De Rosa G, Brugnara C, Alper SL, Snyder LM, Iolascon A. Novel Gardos channel mutations linked to dehydrated hereditary stomatocytosis (xerocytosis). Am J Hematol 2015; 90:921-6. [PMID: 26178367 DOI: 10.1002/ajh.24117] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 07/08/2015] [Indexed: 02/06/2023]
Abstract
Dehydrated hereditary stomatocytosis (DHSt) is an autosomal dominant congenital hemolytic anemia with moderate splenomegaly and often compensated hemolysis. Affected red cells are characterized by a nonspecific cation leak of the red cell membrane, reflected in elevated sodium content, decreased potassium content, elevated MCHC and MCV, and decreased osmotic fragility. The majority of symptomatic DHSt cases reported to date have been associated with gain-of-function mutations in the mechanosensitive cation channel gene, PIEZO1. A recent study has identified two families with DHSt associated with a single mutation in the KCNN4 gene encoding the Gardos channel (KCa3.1), the erythroid Ca(2+) -sensitive K(+) channel of intermediate conductance, also expressed in many other cell types. We present here, in the second report of DHSt associated with KCNN4 mutations, two previously undiagnosed DHSt families. Family NA exhibited the same de novo missense mutation as that recently described, suggesting a hot spot codon for DHSt mutations. Family WO carried a novel, inherited missense mutation in the ion transport domain of the channel. The patients' mild hemolytic anemia did not improve post-splenectomy, but splenectomy led to no serious thromboembolic events. We further characterized the expression of KCNN4 in the mutated patients and during erythroid differentiation of CD34+ cells and K562 cells. We also analyzed KCNN4 expression during mouse embryonic development.
Collapse
Affiliation(s)
- Immacolata Andolfo
- Department Of Molecular Medicine And Medical Biotechnologies; “Federico II” University Of Naples; Naples Italy
- Biotecnologie Avanzate; CEINGE; Naples Italy
| | - Roberta Russo
- Department Of Molecular Medicine And Medical Biotechnologies; “Federico II” University Of Naples; Naples Italy
- Biotecnologie Avanzate; CEINGE; Naples Italy
| | - Francesco Manna
- Department Of Molecular Medicine And Medical Biotechnologies; “Federico II” University Of Naples; Naples Italy
- Biotecnologie Avanzate; CEINGE; Naples Italy
| | - Boris E. Shmukler
- Renal Division And Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School; Boston Massachusetts
- Department Of Medicine, Harvard Medical School; Boston Massachusetts
| | - Antonella Gambale
- Department Of Molecular Medicine And Medical Biotechnologies; “Federico II” University Of Naples; Naples Italy
- Biotecnologie Avanzate; CEINGE; Naples Italy
| | - Giuseppina Vitiello
- Biotecnologie Avanzate; CEINGE; Naples Italy
- Medical Genetics Unit; Policlinico Tor Vergata University Hospital; Viale Oxford Rome Italy
| | - Gianluca De Rosa
- Department Of Molecular Medicine And Medical Biotechnologies; “Federico II” University Of Naples; Naples Italy
- Biotecnologie Avanzate; CEINGE; Naples Italy
| | - Carlo Brugnara
- Department Of Laboratory Medicine, Boston Children's Hospital And Department Of Pathology; Harvard Medical School; Boston Massachusetts
| | - Seth L. Alper
- Renal Division And Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School; Boston Massachusetts
| | - L. Michael Snyder
- Dept Of Hospital Laboratories; University Of Massachusetts Medical Center; Worcester MA
- Quest Diagnositics, LLC MA; Marlborough Massachusetts
| | - Achille Iolascon
- Department Of Molecular Medicine And Medical Biotechnologies; “Federico II” University Of Naples; Naples Italy
- Biotecnologie Avanzate; CEINGE; Naples Italy
| |
Collapse
|
125
|
Architecture of the mammalian mechanosensitive Piezo1 channel. Nature 2015; 527:64-9. [PMID: 26390154 DOI: 10.1038/nature15247] [Citation(s) in RCA: 335] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/24/2015] [Indexed: 12/12/2022]
Abstract
Piezo proteins are evolutionarily conserved and functionally diverse mechanosensitive cation channels. However, the overall structural architecture and gating mechanisms of Piezo channels have remained unknown. Here we determine the cryo-electron microscopy structure of the full-length (2,547 amino acids) mouse Piezo1 (Piezo1) at a resolution of 4.8 Å. Piezo1 forms a trimeric propeller-like structure (about 900 kilodalton), with the extracellular domains resembling three distal blades and a central cap. The transmembrane region has 14 apparently resolved segments per subunit. These segments form three peripheral wings and a central pore module that encloses a potential ion-conducting pore. The rather flexible extracellular blade domains are connected to the central intracellular domain by three long beam-like structures. This trimeric architecture suggests that Piezo1 may use its peripheral regions as force sensors to gate the central ion-conducting pore.
Collapse
|
126
|
Salem JC, Reviriego-Mendoza MM, Santy LC. ARF-GEF cytohesin-2/ARNO regulates R-Ras and α5-integrin recycling through an EHD1-positive compartment. Mol Biol Cell 2015; 26:4265-79. [PMID: 26378252 PMCID: PMC4642859 DOI: 10.1091/mbc.e15-05-0278] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/11/2015] [Indexed: 11/30/2022] Open
Abstract
R-Ras and cytohesin-2/ARNO coordinate in the control of epithelial cell adhesion, but the mechanism has been unclear. Cytohesin-2/ARNO regulates traffic through an EHD1-positive recycling compartment. Inhibition of cytohesin-2/ARNO activity traps R-Ras and integrins within the EHD1 compartment and impairs adhesion and spreading. When expressed in epithelial cells, cytohesin-2/ARNO, a guanine nucleotide exchange factor (GEF) for ARF small GTPases, causes a robust migration response. Recent evidence suggests that cytohesin-2/ARNO acts downstream of small the GTPase R-Ras to promote spreading and migration. We hypothesized that cytohesin-2/ARNO could transmit R-Ras signals by regulating the recycling of R-Ras through ARF activation. We found that Eps15-homology domain 1 (EHD1), a protein that associates with the endocytic recycling compartment (ERC), colocalizes with active R-Ras in transiently expressed HeLa cells. In addition, we show that EHD1-positive recycling endosomes are a novel compartment for cytohesin-2/ARNO. Knockdown or expression of GEF-inactive (E156K) cytohesin-2/ARNO causes R-Ras to accumulate on recycling endosomes containing EHD1 and inhibits cell spreading. E156K-ARNO also causes a reduction in focal adhesion size and number. Finally, we demonstrate that R-Ras/ARNO signaling is required for recycling of α5-integrin and R-Ras to the plasma membrane. These data establish a role for cytohesin-2/ARNO as a regulator of R-Ras and integrin recycling and suggest that ARF-regulated trafficking of R-Ras is required for R-Ras–dependent effects on spreading and adhesion formation.
Collapse
Affiliation(s)
- Joseph C Salem
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
| | - Marta M Reviriego-Mendoza
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
| | - Lorraine C Santy
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
127
|
Fotiou E, Martin-Almedina S, Simpson MA, Lin S, Gordon K, Brice G, Atton G, Jeffery I, Rees DC, Mignot C, Vogt J, Homfray T, Snyder MP, Rockson SG, Jeffery S, Mortimer PS, Mansour S, Ostergaard P. Novel mutations in PIEZO1 cause an autosomal recessive generalized lymphatic dysplasia with non-immune hydrops fetalis. Nat Commun 2015; 6:8085. [PMID: 26333996 PMCID: PMC4568316 DOI: 10.1038/ncomms9085] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/15/2015] [Indexed: 12/19/2022] Open
Abstract
Generalized lymphatic dysplasia (GLD) is a rare form of primary lymphoedema characterized by a uniform, widespread lymphoedema affecting all segments of the body, with systemic involvement such as intestinal and/or pulmonary lymphangiectasia, pleural effusions, chylothoraces and/or pericardial effusions. This may present prenatally as non-immune hydrops. Here we report homozygous and compound heterozygous mutations in PIEZO1, resulting in an autosomal recessive form of GLD with a high incidence of non-immune hydrops fetalis and childhood onset of facial and four limb lymphoedema. Mutations in PIEZO1, which encodes a mechanically activated ion channel, have been reported with autosomal dominant dehydrated hereditary stomatocytosis and non-immune hydrops of unknown aetiology. Besides its role in red blood cells, our findings indicate that PIEZO1 is also involved in the development of lymphatic structures. Primary lymphoedema can lead to the swelling of the extremities and facial dysmorphism. Here the authors present evidence that compound heterozygous and homozygous mutations in PIEZO1 result in an autosomal recessive form of generalised lymphatic dysplasia.
Collapse
Affiliation(s)
- Elisavet Fotiou
- Cardiovascular and Cell Sciences Institute, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Silvia Martin-Almedina
- Cardiovascular and Cell Sciences Institute, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Michael A Simpson
- Department of Medical and Molecular Genetics, Division of Genetics and Molecular Medicine, Kings College London School of Medicine, Guy's Hospital, London SE1 9RY, UK
| | - Shin Lin
- Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305, USA.,Department of Genetics, Stanford University, Stanford, California 94305, USA
| | - Kristiana Gordon
- Department of Dermatology, St. George's Healthcare NHS Trust, London SW17 0QT, UK
| | - Glen Brice
- South West Thames Regional Genetics Unit, St. George's University of London, London SW17 0RE, UK
| | - Giles Atton
- South West Thames Regional Genetics Unit, St. George's University of London, London SW17 0RE, UK
| | - Iona Jeffery
- Pathology Department, St. George's University of London, London SW17 0RE, UK
| | - David C Rees
- Department of Haematological Medicine, King's College London School of Medicine, King's College Hospital, London SE5 9RS, UK
| | - Cyril Mignot
- Département de Génétique, APHP, GH Pitié-Salpêtrière, Centre de Référence des Déficiences Intellectuelles de Causes Rares, 75013 Paris, France
| | - Julie Vogt
- West Midlands Regional Genetics Service, Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham B15 2TG, UK
| | - Tessa Homfray
- South West Thames Regional Genetics Unit, St. George's University of London, London SW17 0RE, UK
| | - Michael P Snyder
- Department of Genetics, Stanford University, Stanford, California 94305, USA
| | - Stanley G Rockson
- Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305, USA
| | - Steve Jeffery
- Cardiovascular and Cell Sciences Institute, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Peter S Mortimer
- Cardiovascular and Cell Sciences Institute, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Sahar Mansour
- South West Thames Regional Genetics Unit, St. George's University of London, London SW17 0RE, UK
| | - Pia Ostergaard
- Cardiovascular and Cell Sciences Institute, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
| |
Collapse
|
128
|
Choi HJ, Sun D, Jakobs TC. Astrocytes in the optic nerve head express putative mechanosensitive channels. Mol Vis 2015; 21:749-66. [PMID: 26236150 PMCID: PMC4502055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/12/2015] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To establish whether optic nerve head astrocytes express candidate molecules to sense tissue stretch. METHODS We used conventional PCR, quantitative PCR, and single-cell reverse transcription PCR (RT-PCR) to assess the expression of various members of the transient receptor potential (TRP) channel family and of the recently characterized mechanosensitive channels Piezo1 and 2 in optic nerve head tissue and in single, isolated astrocytes. RESULTS Most TRP subfamilies (TRPC, TRPM, TRPV, TRPA, and TRPP) and Piezo1 and 2 were expressed in the optic nerve head of the mouse. Quantitative real-time PCR analysis showed that TRPC1, TRPM7, TRPV2, TRPP2, and Piezo1 are the dominant isoforms in each subfamily. Single-cell RT-PCR revealed that many TRP isoforms, TRPC1-2, TRPC6, TRPV2, TRPV4, TRPM2, TRPM4, TRPM6-7, TRPP1-2, and Piezo1-2, are expressed in astrocytes of the optic nerve head, and that most astrocytes express TRPC1 and TRPP1-2. Comparisons of the TRPP and Piezo expression levels between different tissue regions showed that Piezo2 expression was higher in the optic nerve head and the optic nerve proper than in the brain and the corpus callosum. TRPP2 also showed higher expression in the optic nerve head. CONCLUSIONS Astrocytes in the optic nerve head express multiple putative mechanosensitive channels, in particular the recently identified channels Piezo1 and 2. The expression of putative mechanosensitive channels in these cells may contribute to their responsiveness to traumatic or glaucomatous injury.
Collapse
|
129
|
Affiliation(s)
- Jing Li
- a School of Medicine ; University of Leeds ; Leeds , UK
| | | | | |
Collapse
|
130
|
Abstract
Neuronal growth cones are exquisite sensory-motor machines capable of transducing features contacted in their local extracellular environment into guided process extension during development. Extensive research has shown that chemical ligands activate cell surface receptors on growth cones leading to intracellular signals that direct cytoskeletal changes. However, the environment also provides mechanical support for growth cone adhesion and traction forces that stabilize leading edge protrusions. Interestingly, recent work suggests that both the mechanical properties of the environment and mechanical forces generated within growth cones influence axon guidance. In this review we discuss novel molecular mechanisms involved in growth cone force production and detection, and speculate how these processes may be necessary for the development of proper neuronal morphogenesis.
Collapse
Affiliation(s)
- Patrick C Kerstein
- Neuroscience Training Program, Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison Madison, WI, USA
| | - Robert H Nichol
- Neuroscience Training Program, Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison Madison, WI, USA
| | - Timothy M Gomez
- Neuroscience Training Program, Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison Madison, WI, USA
| |
Collapse
|
131
|
Sackmann E. How actin/myosin crosstalks guide the adhesion, locomotion and polarization of cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3132-42. [PMID: 26119326 DOI: 10.1016/j.bbamcr.2015.06.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 01/09/2023]
Abstract
Cell-tissue-tissue interaction is determined by specific short range forces between cell adhesion molecules (CAMs) and ligands of the tissue, long range repulsion forces mediated by cell surface grafted macromolecules and adhesion-induced elastic stresses in the cell envelope. This interplay of forces triggers the rapid random clustering of tightly coupled linkers. By coupling of actin gel patches to the intracellular domains of the CAMs, these clusters can grow in a secondary process resulting in the formation of functional adhesion microdomains (ADs). The ADs can act as biochemical steering centers by recruiting and activating functional proteins, such as GTPases and associated regulating proteins, through electrostatic-hydrophobic forces with cationic lipid domains that act as attractive centers. First, I summarize physical concepts of cell adhesion revealed by studies of biomimetic systems. Then I describe the role of the adhesion domains as biochemical signaling platforms and force transmission centers promoting cellular protrusions, in terms of a shell string model of cells. Protrusion forces are generated by actin gelation triggered by molecular machines (focal adhesion kinase (FAK), Src-kinases and associated adaptors) which assemble around newly formed integrin clusters. They recruit and activate the GTPases Rac-1 and actin gelation promoters to charged membrane domains via electrostatic-hydrophobic forces. The cell front is pushed forward in a cyclic and stepwise manner and the step-width is determined by the dynamics antagonistic interplay between Rac-1 and RhoA. The global cell polarization in the direction of motion is mediated by the actin-microtubule (MT) crosstalk at adhesion domains. Supramolecular actin-MT assemblies at the front help to promote actin polymerization. At the rear they regulate the dismantling of the ADs through the Ca(++)-mediated activation of the protease calpain and trigger their disruption by RhoA mediated contraction via stress fibers. This article is part of a Special Issue entitled: Mechanobiology.
Collapse
Affiliation(s)
- Erich Sackmann
- Technical University Munich, Germany; Physics Department E22/E27, James Franck Str., D85747 Garching, Germany.
| |
Collapse
|
132
|
Yang C, Zhang X, Guo Y, Meng F, Sachs F, Guo J. Mechanical dynamics in live cells and fluorescence-based force/tension sensors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1889-904. [PMID: 25958335 DOI: 10.1016/j.bbamcr.2015.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 04/07/2015] [Accepted: 05/01/2015] [Indexed: 01/13/2023]
Abstract
Three signaling systems play the fundamental roles in modulating cell activities: chemical, electrical, and mechanical. While the former two are well studied, the mechanical signaling system is still elusive because of the lack of methods to measure structural forces in real time at cellular and subcellular levels. Indeed, almost all biological processes are responsive to modulation by mechanical forces that trigger dispersive downstream electrical and biochemical pathways. Communication among the three systems is essential to make cells and tissues receptive to environmental changes. Cells have evolved many sophisticated mechanisms for the generation, perception and transduction of mechanical forces, including motor proteins and mechanosensors. In this review, we introduce some background information about mechanical dynamics in live cells, including the ubiquitous mechanical activity, various types of mechanical stimuli exerted on cells and the different mechanosensors. We also summarize recent results obtained using genetically encoded FRET (fluorescence resonance energy transfer)-based force/tension sensors; a new technique used to measure mechanical forces in structural proteins. The sensors have been incorporated into many specific structural proteins and have measured the force gradients in real time within live cells, tissues, and animals.
Collapse
Affiliation(s)
- Chao Yang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - Xiaohan Zhang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - Yichen Guo
- The University of Alabama, Tuscaloosa, AL, 35401, USA
| | - Fanjie Meng
- Physiology and Biophysics Department, Center for Single Molecule Studies, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Frederick Sachs
- Physiology and Biophysics Department, Center for Single Molecule Studies, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Jun Guo
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China.
| |
Collapse
|
133
|
Caburet S, Anttonen M, Todeschini AL, Unkila-Kallio L, Mestivier D, Butzow R, Veitia RA. Combined comparative genomic hybridization and transcriptomic analyses of ovarian granulosa cell tumors point to novel candidate driver genes. BMC Cancer 2015; 15:251. [PMID: 25884336 PMCID: PMC4407711 DOI: 10.1186/s12885-015-1283-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 03/27/2015] [Indexed: 12/23/2022] Open
Abstract
Background Ovarian granulosa cell tumors (GCTs) are the most frequent sex cord-stromal tumors. Several studies have shown that a somatic mutation leading to a C134W substitution in the transcription factor FOXL2 appears in more than 95% of adult-type GCTs. Its pervasive presence suggests that FOXL2 is the main cancer driver gene. However, other mutations and genomic changes might also contribute to tumor formation and/or progression. Methods We have performed a combined comparative genomic hybridization and transcriptomic analyses of 10 adult-type GCTs to obtain a picture of the genomic landscape of this cancer type and to identify new candidate co-driver genes. Results Our results, along with a review of previous molecular studies, show the existence of highly recurrent chromosomal imbalances (especially, trisomy 14 and monosomy 22) and preferential co-occurrences (i.e. trisomy 14/monosomy 22 and trisomy 7/monosomy 16q). In-depth analyses showed the presence of recurrently broken, amplified/duplicated or deleted genes. Many of these genes, such as AKT1, RUNX1 and LIMA1, are known to be involved in cancer and related processes. Further genomic explorations suggest that they are functionally related. Conclusions Our combined analysis identifies potential candidate genes, whose alterations might contribute to adult-type GCT formation/progression together with the recurrent FOXL2 somatic mutation. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1283-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sandrine Caburet
- Institut Jacques Monod, Paris, France. .,Université Paris Diderot/Paris, Paris, France. .,Université Paris-Diderot & Institut Jacques Monod, CNRS-UMR 7592, Bâtiment Buffon, 15 Rue Hélène Brion, Paris, Cedex 13, France.
| | - Mikko Anttonen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland. .,Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Anne-Laure Todeschini
- Institut Jacques Monod, Paris, France. .,Université Paris Diderot/Paris, Paris, France.
| | - Leila Unkila-Kallio
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Denis Mestivier
- Institut Jacques Monod, Paris, France. .,Université Paris Diderot/Paris, Paris, France.
| | - Ralf Butzow
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland. .,Department of pathology, University of Helsinki, and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland.
| | - Reiner A Veitia
- Institut Jacques Monod, Paris, France. .,Université Paris Diderot/Paris, Paris, France. .,Université Paris-Diderot & Institut Jacques Monod, CNRS-UMR 7592, Bâtiment Buffon, 15 Rue Hélène Brion, Paris, Cedex 13, France.
| |
Collapse
|
134
|
Li C, Rezania S, Kammerer S, Sokolowski A, Devaney T, Gorischek A, Jahn S, Hackl H, Groschner K, Windpassinger C, Malle E, Bauernhofer T, Schreibmayer W. Piezo1 forms mechanosensitive ion channels in the human MCF-7 breast cancer cell line. Sci Rep 2015; 5:8364. [PMID: 25666479 PMCID: PMC4322926 DOI: 10.1038/srep08364] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/23/2014] [Indexed: 01/30/2023] Open
Abstract
Mechanical interaction between cells - specifically distortion of tensional homeostasis-emerged as an important aspect of breast cancer genesis and progression. We investigated the biophysical characteristics of mechanosensitive ion channels (MSCs) in the malignant MCF-7 breast cancer cell line. MSCs turned out to be the most abundant ion channel species and could be activated by negative pressure at the outer side of the cell membrane in a saturable manner. Assessing single channel conductance (GΛ) for different monovalent cations revealed an increase in the succession: Li(+) < Na(+) < K(+) ≈Rb(+) ≈ Cs(+). Divalent cations permeated also with the order: Ca(2+) < Ba(2+). Comparison of biophysical properties enabled us to identify MSCs in MCF-7 as ion channels formed by the Piezo1 protein. Using patch clamp technique no functional MSCs were observed in the benign MCF-10A mammary epithelial cell line. Blocking of MSCs by GsMTx-4 resulted in decreased motility of MCF-7, but not of MCF-10A cells, underscoring a possible role of Piezo1 in invasion and metastatic propagation. The role of Piezo1 in biology and progression of breast cancer is further substantiated by markedly reduced overall survival in patients with increased Piezo1 mRNA levels in the primary tumor.
Collapse
Affiliation(s)
- Chouyang Li
- Department of Biophysics, Medical University of Graz, Graz, Austria
| | - Simin Rezania
- Department of Biophysics, Medical University of Graz, Graz, Austria
| | - Sarah Kammerer
- Department of Biophysics, Medical University of Graz, Graz, Austria
| | - Armin Sokolowski
- Department of Biophysics, Medical University of Graz, Graz, Austria
| | - Trevor Devaney
- Department of Biophysics, Medical University of Graz, Graz, Austria
| | - Astrid Gorischek
- Department of Biophysics, Medical University of Graz, Graz, Austria
| | - Stephan Jahn
- Department of Pathology, Medical University of Graz, Graz, Austria
| | - Hubert Hackl
- Division of Bioinformatics, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Klaus Groschner
- Department of Biophysics, Medical University of Graz, Graz, Austria
| | | | - Ernst Malle
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Thomas Bauernhofer
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | |
Collapse
|
135
|
Delcourt N, Quevedo C, Nonne C, Fons P, O'Brien D, Loyaux D, Diez M, Autelitano F, Guillemot JC, Ferrara P, Muriana A, Callol C, Hérault JP, Herbert JM, Favre G, Bono F. Targeted identification of sialoglycoproteins in hypoxic endothelial cells and validation in zebrafish reveal roles for proteins in angiogenesis. J Biol Chem 2015; 290:3405-17. [PMID: 25384978 PMCID: PMC4319010 DOI: 10.1074/jbc.m114.618611] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Indexed: 11/06/2022] Open
Abstract
The formation of new vessels in the tumor, termed angiogenesis, is essential for primary tumor growth and facilitates tumor invasion and metastasis. Hypoxia has been described as one trigger of angiogenesis. Indeed, hypoxia, which is characterized by areas of low oxygen levels, is a hallmark of solid tumors arising from an imbalance between oxygen delivery and consumption. Hypoxic conditions have profound effects on the different components of the tumoral environment. For example, hypoxia is able to activate endothelial cells, leading to angiogenesis but also thereby initiating a cascade of reactions involving neutrophils, smooth muscle cells, and fibroblasts. In addition, hypoxia directly regulates the expression of many genes for which the role and the importance in the tumoral environment remain to be completely elucidated. In this study, we used a method to selectively label sialoglycoproteins to identify new membrane and secreted proteins involved in the adaptative process of endothelial cells by mass spectrometry-based proteomics. We used an in vitro assay under hypoxic condition to observe an increase of protein expression or modifications of glycosylation. Then the function of the identified proteins was assessed in a vasculogenesis assay in vivo by using a morpholino strategy in zebrafish. First, our approach was validated by the identification of sialoglycoproteins such as CD105, neuropilin-1, and CLEC14A, which have already been described as playing key roles in angiogenesis. Second, we identified several new proteins regulated by hypoxia and demonstrated for the first time the pivotal role of GLUT-1, TMEM16F, and SDF4 in angiogenesis.
Collapse
Affiliation(s)
- Nicolas Delcourt
- From Sanofi Research and Development, 195 route d'Espagne, 31000 Toulouse, France, Centre de recherche en Cancérologie de Toulouse, INSERM UMR1037, Université de Toulouse, 20-24 rue du pont Saint-Pierre, 31057 Toulouse, France
| | - Celia Quevedo
- Biobide, S. L., Paseo Mikeletegi 58, 20009 San Sebastián-Donostia, Spain, and BBD-BioPhenix S. L.-Bionaturis group, Paseo Mikeletegi 56, 20009 San Sebastián-Donostia, Spain
| | - Christelle Nonne
- From Sanofi Research and Development, 195 route d'Espagne, 31000 Toulouse, France
| | - Pierre Fons
- From Sanofi Research and Development, 195 route d'Espagne, 31000 Toulouse, France
| | - Donogh O'Brien
- Donogh O'Brien BioConsulting, Les Poirioux, 18310 St. Outrille, France
| | - Denis Loyaux
- From Sanofi Research and Development, 195 route d'Espagne, 31000 Toulouse, France
| | - Maria Diez
- Biobide, S. L., Paseo Mikeletegi 58, 20009 San Sebastián-Donostia, Spain, and
| | - François Autelitano
- From Sanofi Research and Development, 195 route d'Espagne, 31000 Toulouse, France
| | | | - Pascual Ferrara
- From Sanofi Research and Development, 195 route d'Espagne, 31000 Toulouse, France
| | - Arantza Muriana
- BBD-BioPhenix S. L.-Bionaturis group, Paseo Mikeletegi 56, 20009 San Sebastián-Donostia, Spain
| | - Carlos Callol
- Biobide, S. L., Paseo Mikeletegi 58, 20009 San Sebastián-Donostia, Spain, and
| | - Jean-Pascal Hérault
- From Sanofi Research and Development, 195 route d'Espagne, 31000 Toulouse, France
| | - Jean-Marc Herbert
- From Sanofi Research and Development, 195 route d'Espagne, 31000 Toulouse, France
| | - Gilles Favre
- Centre de recherche en Cancérologie de Toulouse, INSERM UMR1037, Université de Toulouse, 20-24 rue du pont Saint-Pierre, 31057 Toulouse, France
| | - Françoise Bono
- From Sanofi Research and Development, 195 route d'Espagne, 31000 Toulouse, France,
| |
Collapse
|
136
|
Bai T, Qian W, Zhang L, Wang H, Hou XH, Song J. Bioinformatics analysis of Piezo1 and detection of its expression in the gut. Shijie Huaren Xiaohua Zazhi 2015; 23:4816. [DOI: 10.11569/wcjd.v23.i30.4816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
137
|
Li J, Hou B, Tumova S, Muraki K, Bruns A, Ludlow MJ, Sedo A, Hyman AJ, McKeown L, Young RS, Yuldasheva NY, Majeed Y, Wilson LA, Rode B, Bailey MA, Kim HR, Fu Z, Carter DAL, Bilton J, Imrie H, Ajuh P, Dear TN, Cubbon RM, Kearney MT, Prasad RK, Evans PC, Ainscough JFX, Beech DJ. Piezo1 integration of vascular architecture with physiological force. Nature 2014; 515:279-282. [PMID: 25119035 PMCID: PMC4230887 DOI: 10.1038/nature13701] [Citation(s) in RCA: 801] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 07/23/2014] [Indexed: 12/23/2022]
Abstract
The mechanisms by which physical forces regulate endothelial cells to determine the complexities of vascular structure and function are enigmatic. Studies of sensory neurons have suggested Piezo proteins as subunits of Ca(2+)-permeable non-selective cationic channels for detection of noxious mechanical impact. Here we show Piezo1 (Fam38a) channels as sensors of frictional force (shear stress) and determinants of vascular structure in both development and adult physiology. Global or endothelial-specific disruption of mouse Piezo1 profoundly disturbed the developing vasculature and was embryonic lethal within days of the heart beating. Haploinsufficiency was not lethal but endothelial abnormality was detected in mature vessels. The importance of Piezo1 channels as sensors of blood flow was shown by Piezo1 dependence of shear-stress-evoked ionic current and calcium influx in endothelial cells and the ability of exogenous Piezo1 to confer sensitivity to shear stress on otherwise resistant cells. Downstream of this calcium influx there was protease activation and spatial reorganization of endothelial cells to the polarity of the applied force. The data suggest that Piezo1 channels function as pivotal integrators in vascular biology.
Collapse
Affiliation(s)
- Jing Li
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Bing Hou
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Sarka Tumova
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Katsuhiko Muraki
- School of Pharmacy, Aichi-Gakuin University, 1-100 Kusumoto, Chikusa, Nagoya 464-8650, Japan
| | - Alexander Bruns
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Melanie J Ludlow
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Alicia Sedo
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Adam J Hyman
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Lynn McKeown
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Richard S Young
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
- Department of Hepatobiliary and Transplant Surgery, St. James’s University Hospital, Leeds, UK
| | - Nadira Y Yuldasheva
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Yasser Majeed
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Lesley A Wilson
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Baptiste Rode
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Marc A Bailey
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Hyejeong R Kim
- Cardiovascular Science, University of Sheffield, Sheffield, S10 2RX, UK
| | - Zhaojun Fu
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Deborah AL Carter
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Jan Bilton
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Helen Imrie
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Paul Ajuh
- Dundee Cell Products Ltd, James Lindsay Place, Dundee, DD1 5JJ, UK
| | - T Neil Dear
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Richard M Cubbon
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Mark T Kearney
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - Raj K Prasad
- Department of Hepatobiliary and Transplant Surgery, St. James’s University Hospital, Leeds, UK
| | - Paul C Evans
- Cardiovascular Science, University of Sheffield, Sheffield, S10 2RX, UK
| | - Justin FX Ainscough
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| | - David J Beech
- School of Medicine and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
138
|
Weber-Boyvat M, Kentala H, Lilja J, Vihervaara T, Hanninen R, Zhou Y, Peränen J, Nyman TA, Ivaska J, Olkkonen VM. OSBP-related protein 3 (ORP3) coupling with VAMP-associated protein A regulates R-Ras activity. Exp Cell Res 2014; 331:278-91. [PMID: 25447204 DOI: 10.1016/j.yexcr.2014.10.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 12/20/2022]
Abstract
ORP3 is an R-Ras interacting oxysterol-binding protein homolog that regulates cell adhesion and is overexpressed in several cancers. We investigated here a novel function of ORP3 dependent on its targeting to both the endoplasmic reticulum (ER) and the plasma membrane (PM). Using biochemical and cell imaging techniques we demonstrate the mechanistic requirements for the subcellular targeting and function of ORP3 in control of R-Ras activity. We show that hyperphosphorylated ORP3 (ORP3-P) selectively interacts with the ER membrane protein VAPA, and ORP3-VAPA complexes are targeted to PM sites via the ORP3 pleckstrin homology (PH) domain. A novel FFAT (two phenylalanines in an acidic tract)-like motif was identified in ORP3; only disruption of both the FFAT-like and canonical FFAT motif abolished the phorbol-12-myristate-13-acetate (PMA) stimulated interaction of ORP3-P with VAPA. Co-expression of ORP3 and VAPA induced R-Ras activation, dependent on the interactions of ORP3 with VAPA and the PM. Consistently, downstream AktS473 phosphorylation and β1-integrin activity were enhanced by ORP3-VAPA. To conclude, phosphorylation of ORP3 controls its association with VAPA. Furthermore, we present evidence that ORP3-VAPA complexes stimulate R-Ras signaling.
Collapse
Affiliation(s)
- Marion Weber-Boyvat
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland
| | - Henriikka Kentala
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland
| | - Johanna Lilja
- VTT Medical Biotechnology and Turku Centre for Biotechnology, University of Turku, FI-20520 Turku, Finland
| | - Terhi Vihervaara
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland
| | - Raisa Hanninen
- National Institute for Health and Welfare, Biomedicum 1, FI-00290 Helsinki, Finland
| | - You Zhou
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland
| | - Johan Peränen
- Institute of Biotechnology, FI-00014 University of Helsinki, Finland
| | - Tuula A Nyman
- Institute of Biotechnology, FI-00014 University of Helsinki, Finland
| | - Johanna Ivaska
- VTT Medical Biotechnology and Turku Centre for Biotechnology, University of Turku, FI-20520 Turku, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland; Institute of Biomedicine/Anatomy, FI-00014 University of Helsinki, Finland.
| |
Collapse
|
139
|
Piezo channels: from structure to function. Pflugers Arch 2014; 467:95-9. [DOI: 10.1007/s00424-014-1578-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/03/2014] [Accepted: 07/08/2014] [Indexed: 10/25/2022]
|
140
|
Yang XN, Lu YP, Liu JJ, Huang JK, Liu YP, Xiao CX, Jazag A, Ren JL, Guleng B. Piezo1 is as a novel trefoil factor family 1 binding protein that promotes gastric cancer cell mobility in vitro. Dig Dis Sci 2014; 59:1428-35. [PMID: 24798994 DOI: 10.1007/s10620-014-3044-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 01/20/2014] [Indexed: 12/09/2022]
Abstract
BACKGROUND Trefoil factor family 1 (TFF1) is a member of the TFF-domain peptide family involved in epithelial restitution and cell motility. Recently, we screened Piezo1 as a candidate TFF1-binding protein. AIM We aimed to confirm Piezo1 as a novel TFF1 binding protein and to assess the role of this interaction in mediating gastric cancer cell mobility. METHODS This interaction was confirmed by co-immunoprecipitation and co-localisation of TFF1 and Piezo1 in GES-1 cells. We used stable RNA interference to knockdown Piezo1 protein expression and restored the expression of TFF1 in the gastric cancer cell lines SGC-7901 and BGC-823. Cell motility was evaluated using invasion assay and migration assay in vitro. The expression levels of the integrin subunits β1, β5, α1 as well as the expression of β-catenin and E-cadherin were detected by Western blot. RESULTS We demonstrate that TFF1, but not TFF2 or TFF3, bind to and co-localize with Piezo1 in the cytoplasm in vitro. TFF1 interacts with the C-terminal portion of the Piezo1 protein. Wound healing and trans-well assays demonstrated that the restored expression of TFF1 promoted cell mobility in gastric cancer cells, and this effect was attenuated by the knockdown of Piezo1. Western blots demonstrated the decreased expression of integrin β1 in Piezo1-knockdown cells. CONCLUSIONS Our data demonstrate that Piezo1 is a novel TFF1 binding protein that is important for TFF1-mediated cell migration and suggest that this interaction may be a therapeutic target in the invasion and metastasis of gastric cancer.
Collapse
Affiliation(s)
- Xiao-Ning Yang
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Miyamoto T, Mochizuki T, Nakagomi H, Kira S, Watanabe M, Takayama Y, Suzuki Y, Koizumi S, Takeda M, Tominaga M. Functional role for Piezo1 in stretch-evoked Ca²⁺ influx and ATP release in urothelial cell cultures. J Biol Chem 2014; 289:16565-75. [PMID: 24759099 DOI: 10.1074/jbc.m113.528638] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The urothelium is a sensory structure that contributes to mechanosensation in the urinary bladder. Here, we provide evidence for a critical role for the Piezo1 channel, a newly identified mechanosensory molecule, in the mouse bladder urothelium. We performed a systematic analysis of the molecular and functional expression of Piezo1 channels in the urothelium. Immunofluorescence examination demonstrated abundant expression of Piezo1 in the mouse and human urothelium. Urothelial cells isolated from mice exhibited a Piezo1-dependent increase in cytosolic Ca(2+) concentrations in response to mechanical stretch stimuli, leading to potent ATP release; this response was suppressed in Piezo1-knockdown cells. In addition, Piezo1 and TRPV4 distinguished different intensities of mechanical stimulus. Moreover, GsMTx4, an inhibitor of stretch-activated channels, attenuated the Ca(2+) influx into urothelial cells and decreased ATP release from them upon stretch stimulation. These results suggest that Piezo1 senses extension of the bladder urothelium, leading to production of an ATP signal. Thus, inhibition of Piezo1 might provide a promising means of treating bladder dysfunction.
Collapse
Affiliation(s)
- Tatsuya Miyamoto
- From the Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898
| | - Tsutomu Mochizuki
- From the Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898,
| | - Hiroshi Nakagomi
- From the Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898
| | - Satoru Kira
- From the Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898
| | - Masaki Watanabe
- the Division of Cell Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, Aichi 444-8787
| | - Yasunori Takayama
- the Division of Cell Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, Aichi 444-8787
| | - Yoshiro Suzuki
- the Division of Cell Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, Aichi 444-8787, the Department of Physiological Sciences, Graduate University for Advanced Studies, Okazaki 444-8585, and
| | - Schuichi Koizumi
- the Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Masayuki Takeda
- From the Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898
| | - Makoto Tominaga
- the Division of Cell Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, Aichi 444-8787, the Department of Physiological Sciences, Graduate University for Advanced Studies, Okazaki 444-8585, and
| |
Collapse
|
142
|
Sackmann E, Smith AS. Physics of cell adhesion: some lessons from cell-mimetic systems. SOFT MATTER 2014; 10:1644-59. [PMID: 24651316 PMCID: PMC4028615 DOI: 10.1039/c3sm51910d] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cell adhesion is a paradigm of the ubiquitous interplay of cell signalling, modulation of material properties and biological functions of cells. It is controlled by competition of short range attractive forces, medium range repellant forces and the elastic stresses associated with local and global deformation of the composite cell envelopes. We review the basic physical rules governing the physics of cell adhesion learned by studying cell-mimetic systems and demonstrate the importance of these rules in the context of cellular systems. We review how adhesion induced micro-domains couple to the intracellular actin and microtubule networks allowing cells to generate strong forces with a minimum of attractive cell adhesion molecules (CAMs) and to manipulate other cells through filopodia over micrometer distances. The adhesion strength can be adapted to external force fluctuations within seconds by varying the density of attractive and repellant CAMs through exocytosis and endocytosis or protease-mediated dismantling of the CAM-cytoskeleton link. Adhesion domains form local end global biochemical reaction centres enabling the control of enzymes. Actin-microtubule crosstalk at adhesion foci facilitates the mechanical stabilization of polarized cell shapes. Axon growth in tissue is guided by attractive and repulsive clues controlled by antagonistic signalling pathways.
Collapse
Affiliation(s)
- Erich Sackmann
- Physics Department Technical University Munich, Germany
- Department of Physics, Ludwig-Maximillian University, Munich, Germany
| | - Ana-Sunčana Smith
- Institute for Theoretical Physics, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
- Institute Rud̷er Bošković, Zagreb, Croatia.
| |
Collapse
|
143
|
Abstract
Living organisms sense their physical environment through cellular mechanotransduction, which converts mechanical forces into electrical and biochemical signals. In turn, signal transduction serves a wide variety of functions, from basic cellular processes as diverse as proliferation, differentiation, migration, and apoptosis up to some of the most sophisticated senses, including touch and hearing. Accordingly, defects in mechanosensing potentially lead to diverse diseases and disorders such as hearing loss, cardiomyopathies, muscular dystrophies, chronic pain, and cancer. Here, we review the status of mechanically activated ion channel discovery and discuss current challenges to define their properties and physiological functions.
Collapse
Affiliation(s)
- Patrick Delmas
- Aix-Marseille-Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, UMR 7286, CS80011, Bd Pierre Dramard, 13344 Marseille, Cedex 15, France.
| | | |
Collapse
|
144
|
Evensen NA, Kuscu C, Nguyen HL, Zarrabi K, Dufour A, Kadam P, Hu YJ, Pulkoski-Gross A, Bahou WF, Zucker S, Cao J. Unraveling the role of KIAA1199, a novel endoplasmic reticulum protein, in cancer cell migration. J Natl Cancer Inst 2013; 105:1402-16. [PMID: 23990668 DOI: 10.1093/jnci/djt224] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cell migration is a critical determinant of cancer metastasis, and a better understanding of the genes involved will lead to the identification of novel targets aimed at preventing cancer dissemination. KIAA1199 has been shown to be upregulated in human cancers, yet its role in cancer progression was hitherto unknown. METHODS Clinical relevance was assessed by examining KIAA1199 expression in human cancer specimens. In vitro and in vivo studies were employed to determine the function of KIAA1199 in cancer progression. Cellular localization of KIAA1199 was microscopically determined. SNAP-tag pull-down assays were used to identify binding partner(s) of KIAA1199. Calcium levels were evaluated using spectrofluorometric and fluorescence resonance energy transfer analyses. Signaling pathways were dissected by Western blotting. Student t test was used to assess differences. All statistical tests were two-sided. RESULTS KIAA1199 was upregulated in invasive breast cancer specimens and inversely associated with patient survival rate. Silencing of KIAA1199 in MDA-MB-435 cancer cells resulted in a mesenchymal-to-epithelial transition that reduced cell migratory ability in vitro (75% reduction; P < .001) and decreased metastasis in vivo (80% reduction; P < .001). Gain-of-function assays further demonstrated the role of KIAA1199 in cell migration. KIAA1199-enhanced cell migration required endoplasmic reticulum (ER) localization, where it forms a stable complex with the chaperone binding immunoglobulin protein (BiP). A novel ER-retention motif within KIAA1199 that is required for its ER localization, BiP interaction, and enhanced cell migration was identified. Mechanistically, KIAA1199 was found to mediate ER calcium leakage, and the resultant increase in cytosolic calcium ultimately led to protein kinase C alpha activation and cell migration. CONCLUSIONS KIAA1199 serves as a novel cell migration-promoting gene and plays a critical role in maintaining cancer mesenchymal status.
Collapse
Affiliation(s)
- Nikki A Evensen
- Affiliations of authors: Department of Medicine/Cancer Prevention (NAE, CK, H-LN, KZ, AD, AP-G, JC), Department of Pathology (YH, JC), Department of Medicine/Hematology & Oncology (WFB, SZ), Stony Brook University, Stony Brook, NY; Department of Research, Veterans Affair Medical Center, Northport, NY (H-LN, PK, SZ); Centre for Blood Research and Departments of Biochemistry and Molecular Biology and Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC, Canada (AD)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Saraiva N, Prole DL, Carrara G, Johnson BF, Taylor CW, Parsons M, Smith GL. hGAAP promotes cell adhesion and migration via the stimulation of store-operated Ca2+ entry and calpain 2. ACTA ACUST UNITED AC 2013; 202:699-713. [PMID: 23940116 PMCID: PMC3747308 DOI: 10.1083/jcb.201301016] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Golgi antiapoptotic proteins (GAAPs) are highly conserved Golgi membrane proteins that inhibit apoptosis and promote Ca(2+) release from intracellular stores. Given the role of Ca(2+) in controlling cell adhesion and motility, we hypothesized that human GAAP (hGAAP) might influence these events. In this paper, we present evidence that hGAAP increased cell adhesion, spreading, and migration in a manner that depended on the C-terminal domain of hGAAP. We show that hGAAP increased store-operated Ca(2+) entry and thereby the activity of calpain at newly forming protrusions. These hGAAP-dependent effects regulated focal adhesion dynamics and cell migration. Indeed, inhibition or knockdown of calpain 2 abrogated the effects of hGAAP on cell spreading and migration. Our data reveal that hGAAP is a novel regulator of focal adhesion dynamics, cell adhesion, and migration by controlling localized Ca(2+)-dependent activation of calpain.
Collapse
Affiliation(s)
- Nuno Saraiva
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, England, UK
| | | | | | | | | | | | | |
Collapse
|
146
|
Ross TD, Coon BG, Yun S, Baeyens N, Tanaka K, Ouyang M, Schwartz MA. Integrins in mechanotransduction. Curr Opin Cell Biol 2013; 25:613-8. [PMID: 23797029 DOI: 10.1016/j.ceb.2013.05.006] [Citation(s) in RCA: 249] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/31/2013] [Accepted: 05/31/2013] [Indexed: 12/18/2022]
Abstract
Forces acting on cells govern many important regulatory events during development, normal physiology, and disease processes. Integrin-mediated adhesions, which transmit forces between the extracellular matrix and the actin cytoskeleton, play a central role in transducing effects of forces to regulate cell functions. Recent work has led to major insights into the molecular mechanisms by which these adhesions respond to forces to control cellular signaling pathways. We briefly summarize effects of forces on organs, tissues, and cells; and then discuss recent advances toward understanding molecular mechanisms.
Collapse
Affiliation(s)
- Tyler D Ross
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, United States
| | | | | | | | | | | | | |
Collapse
|
147
|
Chen CC, Wong CW. Neurosensory mechanotransduction through acid-sensing ion channels. J Cell Mol Med 2013; 17:337-49. [PMID: 23490035 PMCID: PMC3823015 DOI: 10.1111/jcmm.12025] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 12/28/2012] [Indexed: 02/06/2023] Open
Abstract
Acid-sensing ion channels (ASICs) are voltage-insensitive cation channels responding to extracellular acidification. ASIC proteins have two transmembrane domains and a large extracellular domain. The molecular topology of ASICs is similar to that of the mechanosensory abnormality 4- or 10-proteins expressed in touch receptor neurons and involved in neurosensory mechanotransduction in nematodes. The ASIC proteins are involved in neurosensory mechanotransduction in mammals. The ASIC isoforms are expressed in Merkel cell-neurite complexes, periodontal Ruffini endings and specialized nerve terminals of skin and muscle spindles, so they might participate in mechanosensation. In knockout mouse models, lacking an ASIC isoform produces defects in neurosensory mechanotransduction of tissue such as skin, stomach, colon, aortic arch, venoatrial junction and cochlea. The ASICs are thus implicated in touch, pain, digestive function, baroreception, blood volume control and hearing. However, the role of ASICs in mechanotransduction is still controversial, because we lack evidence that the channels are mechanically sensitive when expressed in heterologous cells. Thus, ASIC channels alone are not sufficient to reconstruct the path of transducing molecules of mechanically activated channels. The mechanotransducers associated with ASICs need further elucidation. In this review, we discuss the expression of ASICs in sensory afferents of mechanoreceptors, findings of knockout studies, technical issues concerning studies of neurosensory mechanotransduction and possible missing links. Also we propose a molecular model and a new approach to disclose the molecular mechanism underlying the neurosensory mechanotransduction.
Collapse
Affiliation(s)
- Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | | |
Collapse
|
148
|
Multiple clinical forms of dehydrated hereditary stomatocytosis arise from mutations in PIEZO1. Blood 2013; 121:3925-35, S1-12. [PMID: 23479567 DOI: 10.1182/blood-2013-02-482489] [Citation(s) in RCA: 237] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Autosomal dominant dehydrated hereditary stomatocytosis (DHSt) usually presents as a compensated hemolytic anemia with macrocytosis and abnormally shaped red blood cells (RBCs). DHSt is part of a pleiotropic syndrome that may also exhibit pseudohyperkalemia and perinatal edema. We identified PIEZO1 as the disease gene for pleiotropic DHSt in a large kindred by exome sequencing analysis within the previously mapped 16q23-q24 interval. In 26 affected individuals among 7 multigenerational DHSt families with the pleiotropic syndrome, 11 heterozygous PIEZO1 missense mutations cosegregated with disease. PIEZO1 is expressed in the plasma membranes of RBCs and its messenger RNA, and protein levels increase during in vitro erythroid differentiation of CD34(+) cells. PIEZO1 is also expressed in liver and bone marrow during human and mouse development. We suggest for the first time a correlation between a PIEZO1 mutation and perinatal edema. DHSt patient red cells with the R2456H mutation exhibit increased ion-channel activity. Functional studies of PIEZO1 mutant R2488Q expressed in Xenopus oocytes demonstrated changes in ion-channel activity consistent with the altered cation content of DHSt patient red cells. Our findings provide direct evidence that R2456H and R2488Q mutations in PIEZO1 alter mechanosensitive channel regulation, leading to increased cation transport in erythroid cells.
Collapse
|
149
|
Albuisson J, Murthy SE, Bandell M, Coste B, Louis-Dit-Picard H, Mathur J, Fénéant-Thibault M, Tertian G, de Jaureguiberry JP, Syfuss PY, Cahalan S, Garçon L, Toutain F, Simon Rohrlich P, Delaunay J, Picard V, Jeunemaitre X, Patapoutian A. Dehydrated hereditary stomatocytosis linked to gain-of-function mutations in mechanically activated PIEZO1 ion channels. Nat Commun 2013; 4:1884. [PMID: 23695678 PMCID: PMC3674779 DOI: 10.1038/ncomms2899] [Citation(s) in RCA: 266] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 04/18/2013] [Indexed: 02/06/2023] Open
Abstract
Dehydrated hereditary stomatocytosis is a genetic condition with defective red blood cell membrane properties that causes an imbalance in intracellular cation concentrations. Recently, two missense mutations in the mechanically activated PIEZO1 (FAM38A) ion channel were associated with dehydrated hereditary stomatocytosis. However, it is not known how these mutations affect PIEZO1 function. Here, by combining linkage analysis and whole-exome sequencing in a large pedigree and Sanger sequencing in two additional kindreds and 11 unrelated dehydrated hereditary stomatocytosis cases, we identify three novel missense mutations and one recurrent duplication in PIEZO1, demonstrating that it is the major gene for dehydrated hereditary stomatocytosis. All the dehydrated hereditary stomatocytosis-associated mutations locate at C-terminal half of PIEZO1. Remarkably, we find that all PIEZO1 mutations give rise to mechanically activated currents that inactivate more slowly than wild-type currents. This gain-of-function PIEZO1 phenotype provides insight that helps to explain the increased permeability of cations in red blood cells of dehydrated hereditary stomatocytosis patients. Our findings also suggest a new role for mechanotransduction in red blood cell biology and pathophysiology.
Collapse
|
150
|
Loss of the integrin-activating transmembrane protein Fam38A (Piezo1) promotes a switch to a reduced integrin-dependent mode of cell migration. PLoS One 2012; 7:e40346. [PMID: 22792288 PMCID: PMC3390408 DOI: 10.1371/journal.pone.0040346] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 06/04/2012] [Indexed: 01/22/2023] Open
Abstract
Lung cancer is one of the most common fatal diseases in the developed world. The disease is rarely cured by currently available therapies, with an overall survival rate of ∼10%. Characterizing novel proteins that offer crucial insights into the processes of lung tumour invasion and metastasis may therefore provide much-needed prognostic markers, and influence therapeutic strategies. Aberrant function of the integrin family of heterodimeric cell surface receptors is a common theme in cancer - investigation into novel integrin activity regulators may offer crucial insights into the processes of tumour invasion and metastasis and may reveal insights into potential therapeutic targets. We previously described that depletion of the novel multi-transmembrane domain protein Fam38A, located at the endoplasmic reticulum (ER), inactivates endogenous beta1 integrin affinity, reducing cell adhesion. We now show that depletion of Fam38A, also now known as Piezo1, causes anchorage independence and a switch to a reduced integrin-dependent mode of cell migration/invasion, a novel phenotype for this integrin-regulating protein. Normal lung epithelial cells show increased rates of migration by 2D time-lapse microscopy and increased capacity to invade into matrigel, despite having decreased integrin affinity. We confirm greatly depleted Fam38A expression in small cell lung cancer (SCLC) lines where a form of reduced integrin-dependent migration, i.e. amoeboid migration, is a known phenotype. We propose that loss of Fam38A expression may cause increased cell migration and metastasis in lung tumours.
Collapse
|