UB Vice President for Research and Economic Development markup
Frederick Sachs UB Profile
Physiology and Biophysics
SUNY Distinguished Professor
301 Cary Hall
SUNY at Buffalo
Buffalo NY 14214
716-829-5161
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Deekshitha Jetta, Philip A. Gottlieb, Deepika Verma, Frederick Sachs, Susan Z. Hua
PubMedId: 31076516
Journal-article: Journal of Cell Science, pp. jcs.226076, 2019
Authors: Mohammad M. Maneshi, Frederick Sachs, Susan Z. Hua
PubMedId: 29615869
Journal-article: Frontiers in Cellular Neuroscience, vol. 12, 2018
Authors: Christopher W. Ward, Frederick Sachs, Ernest D. Bush, Thomas M. Suchyna
PubMedId: 30174173
Journal-article: Neuromuscular Disorders, 2018
Authors: Radhakrishnan Gnanasambandam, Chilman Bae, Lynn Ziegler, Frederick Sachs, Philip A. Gottlieb
PubMedId: 30462693
Journal-article: PLOS ONE, vol. 13, pp. e0207309, 2018
Authors: Geraldine Polido, Xiangyan Shi, Dian Xu, Chengchen Guo, Rich Thai, Joseph P. Patterson, Nathan C. Gianneschi, Thomas M. Suchyna, Frederick Sachs, Gregory P. Holland
PubMedId: 30463698
Journal-article: Biochimica et Biophysica Acta (BBA) - Biomembranes, 2018
Authors: Pei-Chuan Chao, Mettupalayam Sivaselvan, Frederick Sachs
PubMedId: 30243441
1: Current topics in membranes, vol. 81, 2018
Authors: Mohammad Mehdi Maneshi, Bruce Maki, Radhakrishnan Gnanasambandam, Sophie Belin, Gabriela K. Popescu, Frederick Sachs, Susan Z. Hua
Abstract: While studying the physiological response of primary rat astrocytes to fluid shear stress in a model of traumatic brain injury (TBI), we found that shear stress induced Ca2+ entry. The influx was inhibited by MK-801, a specific pore blocker of N-Methyl-D-aspartic acid receptor (NMDAR) channels, and this occurred in the absence of agonists. Other NMDA open channel blockers ketamine and memantine showed a similar effect. The competitive glutamate antagonists AP5 and GluN2B-selective inhibitor ifenprodil reduced NMDA-activated currents, but had no effect on the mechanically induced Ca2+ influx. Extracellular Mg2+ at 2mM did not significantly affect the shear induced Ca2+ influx, but at 10mM it produced significant inhibition. Patch clamp experiments showed mechanical activation of NMDAR and inhibition by MK-801. The mechanical sensitivity of NMDARs may play a role in the normal physiology of fluid flow in the glymphatic system and it has obvious relevance to TBI.
PubMedId: 28045032
Journal-article: Scientific Reports, vol. 7, pp. 39610, 2017
Authors: Radhakrishnan Gnanasambandam, Chiranjib Ghatak, Anthony Yasmann, Kazuhisa Nishizawa, Frederick Sachs, Alexey S. Ladokhin, Sergei I. Sukharev, Thomas M. Suchyna
PubMedId: 28076814
Journal-article: Biophysical Journal, vol. 112, pp. 31-45, 2017
Authors: Deepika Verma, Vivek K. Bajpai, Nannan Ye, Mohammad M. Maneshi, Deekshitha Jetta, Stelios T. Andreadis, Frederick Sachs, Susan Z. Hua
PubMedId: 28803065
Journal-article: Experimental Cell Research, vol. 359, pp. 327-336, 2017
Authors: Frederick Sachs
PubMedId: 29320693
1: Biophysical journal, vol. 114, 2017
Authors: Wenjun Zheng, Frederick Sachs
PubMedId: 28905417
1: Proteins, vol. 85, 2017
Authors: Frederick Sachs
PubMedId: 28078202
1: World journal of neurology, vol. 5, 2017
Authors: Chilman Bae, Thomas M. Suchyna, Lynn Ziegler, Frederick Sachs, Philip A. Gottlieb
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.
PubMedId: 26963637
Journal-article: PLOS ONE, vol. 11, pp. e0151289, 2016
Authors: Jinli Wang, Yina Ma, Frederick Sachs, Ji Li, Thomas M. Suchyna
PubMedId: 27423272
Journal-article: Journal of Molecular and Cellular Cardiology, vol. 98, pp. 83-94, 2016
Authors: Charles D. Cox, Chilman Bae, Lynn Ziegler, Silas Hartley, Vesna Nikolova-Krstevski, Paul R. Rohde, Chai-Ann Ng, Frederick Sachs, Philip A. Gottlieb, Boris Martinac
PubMedId: 26785635
Journal-article: Nature Communications, vol. 7, pp. 10366, 2016
Authors: Radhakrishnan Gnanasambandam, Morten S Nielsen, Christopher Nicolai, Frederick Sachs, Johannes P Hofgaard, Jakob K Dreyer
PubMedId: 28496407
1: Frontiers in neuroinformatics, vol. 11, 2016
Authors: Anthony W Peng, Radhakrishnan Gnanasambandam, Frederick Sachs, Anthony J Ricci
PubMedId: 26961949
1: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 36, 2016
Authors: Radhakrishnan Gnanasambandam, Chilman Bae, Philip A. Gottlieb, Frederick Sachs
Abstract: Members of the eukaryotic PIEZO family (the human orthologs are noted hPIEZO1 and hPIEZO2) form cation-selective mechanically-gated channels. We characterized the selectivity of human PIEZO1 (hPIEZO1) for alkali ions: K+, Na+, Cs+ and Li+; organic cations: TMA and TEA, and divalents: Ba2+, Ca2+, Mg2+ and Mn2+. All monovalent ions permeated the channel. At a membrane potential of -100 mV, Cs+, Na+ and K+ had chord conductances in the range of 35-55 pS with the exception of Li+, which had a significantly lower conductance of ~ 23 pS. The divalents decreased the single-channel permeability of K+, presumably because the divalents permeated slowly and occupied the open channel for a significant fraction of the time. In cell-attached mode, 90 mM extracellular divalents had a conductance for inward currents carried by the divalents of: 25 pS for Ba2+ and 15 pS for Ca2+ at -80 mV and 10 pS for Mg2+ at -50 mV. The organic cations, TMA and TEA, permeated slowly and attenuated K+ currents much like the divalents. As expected, the channel K+ conductance increased with K+ concentration saturating at ~ 45 pS and the KD of K+ for the channel was 32 mM. Pure divalent ion currents were of lower amplitude than those with alkali ions and the channel opening rate was lower in the presence of divalents than in the presence of monovalents. Exposing cells to the actin disrupting reagent cytochalasin D increased the frequency of openings in cell-attached patches probably by reducing mechanoprotection.
PubMedId: 25955826
Journal-article: PLOS ONE, vol. 10, pp. e0125503, 2015
Authors: Chao Yang, Xiaohan Zhang, Yichen Guo, Fanjie Meng, Frederick Sachs, Jun Guo
Keywords: Fluorescence-based force sensors Mechanical dynamics Mechanosensors Motor proteins Structural force
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.
PubMedId: 25958335
Journal-article: Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol. 1853, pp. 1889-1904, 2015
Authors: Mohammad Mehdi Maneshi, Frederick Sachs, Susan Z. Hua
Keywords: blast intracellular Ca2+ mechanosensitive ion channel (MSC) shear stress traumatic brain injury (TBI)
Abstract: Traumatic brain injury (TBI) refers to brain damage resulting from external mechanical force, such as a blast or crash. Our current understanding of TBI is derived mainly from in vivo studies that show measurable biological effects on neurons sampled after TBI. Little is known about the early responses of brain cells during stimuli and which features of the stimulus are most critical to cell injury. We generated defined shear stress in a microfluidic chamber using a fast pressure servo and examined the intracellular Ca(2+) levels in cultured adult astrocytes. Shear stress increased intracellular Ca(2+) depending on the magnitude, duration, and rise time of the stimulus. Square pulses with a fast rise time (2ms) caused transient increases in intracellular Ca(2+), but when the rise time was extended to 20ms, the response was much less. The threshold for a response is a matrix of multiple parameters. Cells can integrate the effect of shear force from repeated challenges: A pulse train of 10 narrow pulses (11.5dyn/cm(2) and 10ms wide) resulted in a 4-fold increase in Ca(2+) relative to a single pulse of the same amplitude 100ms wide. The Ca(2+) increase was eliminated in Ca(2+)-free media, but was observed after depleting the intracellular Ca(2+) stores with thapsigargin suggesting the need for a Ca(2+) influx. The Ca(2+) influx was inhibited by extracellular Gd(3+), a nonspecific inhibitor of mechanosensitive ion channels, but it was not affected by the more specific inhibitor, GsMTx4. The voltage-gated channel blockers, nifedipine, diltiazem, and verapamil, were also ineffective. The data show that the mechanically induced Ca(2+) influx commonly associated with neuron models for TBI is also present in astrocytes, and there is a viscoelastic/plastic coupling of shear stress to the Ca(2+) influx. The site of Ca(2+) influx has yet to be determined.
PubMedId: 25442327
Journal-article: Journal of Neurotrauma, vol. 32, pp. 1020-1029, 2015
Authors: Deepika Verma, Fanjie Meng, Frederick Sachs, Susan Z Hua
Keywords: FRET MDCK cells cytoskeletal force focal adhesions mechanotransduction
Abstract: Cells respond to fluid shear stress through dynamic processes involving changes in actomyosin and other cytoskeletal stresses, remodeling of cell adhesions, and cytoskeleton reorganization. In this study we simultaneously measured focal adhesion dynamics and cytoskeletal stress and reorganization in MDCK cells under fluid shear stress. The measurements used co-expression of fluorescently labeled paxillin and force sensitive FRET probes of -actinin. A shear stress of 0.74 dyn/cm(2) for 3 hours caused redistribution of cytoskeletal tension and significant focal adhesion remodeling. The fate of focal adhesions is determined by the stress state and stability of the linked actin stress fibers. In the interior of the cell, the mature focal adhesions disassembled within 35-40 min under flow and stress fibers disintegrated. Near the cell periphery, the focal adhesions anchoring the stress fibers perpendicular to the cell periphery disassembled, while focal adhesions associated with peripheral fibers sustained. The diminishing focal adhesions are coupled with local cytoskeletal stress release and actin stress fiber disassembly whereas sustaining peripheral focal adhesions are coupled with an increase in stress and enhancement of actin bundles. The results show that flow induced formation of peripheral actin bundles provides a favorable environment for focal adhesion remodeling along the cell periphery. Under such condition, new FAs were observed along the cell edge under flow. Our results suggest that the remodeling of FAs in epithelial cells under flow is orchestrated by actin cytoskeletal stress redistribution and structural reorganization.
PubMedId: 26418333
Journal-article: Cell Adhesion & Migration, vol. 9, pp. 432-440, 2015
Authors: Kazuhisa Nishizawa, Manami Nishizawa, Radhakrishnan Gnanasambandam, Frederick Sachs, Sergei I. Sukharev, Thomas M. Suchyna
PubMedId: 26342676
Journal-article: Biochimica et Biophysica Acta (BBA) - Biomembranes, vol. 1848, pp. 2767-2778, 2015
Authors: Chilman Bae, Frederick Sachs, Philip A. Gottlieb
PubMedId: 25561736
Journal-article: Journal of Biological Chemistry, vol. 290, pp. 5167-5173, 2015
Authors: Frederick Sachs, Ophir Flomenbom
PubMedId: 25890916
1: Physics of life reviews, vol. 13, 2015
Authors: Frederick Sachs, Mettupalayam V Sivaselvan
PubMedId: 25870207
1: The Journal of general physiology, vol. 145, 2015
Authors: Jun Guo, Yuexiu Wang, Frederick Sachs, Fanjie Meng
PubMedId: 25422450
1: Proceedings of the National Academy of Sciences of the United States of America, vol. 111, 2014
Authors: Whasil Lee, Holly A Leddy, Yong Chen, Suk Hee Lee, Nicole A Zelenski, Amy L McNulty, Jason Wu, Kellie N Beicker, Jeffrey Coles, Stefan Zauscher, J?rg Grandl, Frederick Sachs, Farshid Guilak, Wolfgang B Liedtke
PubMedId: 25385580
1: Proceedings of the National Academy of Sciences of the United States of America, vol. 111, 2014
Authors: Huazhi Han, Avia Rosenhouse-Dantsker, Radhakrishnan Gnanasambandam, Yulia Epshtein, Zhenlong Chen, Frederick Sachs, Richard D Minshall, Irena Levitan
PubMedId: 25038242
1: The Journal of physiology, vol. 592, 2014
Authors: Jean Chemin, Amanda Jane Patel, Patrick Delmas, Frederick Sachs, Michel Lazdunski, Eric Honore
PubMedId: 25168137
1: Current topics in membranes, vol. 59, 2014
Authors: Patrick C Kerstein, Bridget T Jacques-Fricke, Juliana Rengifo, Brian J Mogen, Justin C Williams, Philip A Gottlieb, Fredrick Sachs, Timothy M Gomez
Abstract: Intracellular Ca(2+) signals control the development and regeneration of spinal axons downstream of chemical guidance cues, but little is known about the roles of mechanical cues in axon guidance. Here we show that transient receptor potential canonical 1 (TRPC1) subunits assemble mechanosensitive (MS) channels on Xenopus neuronal growth cones that regulate the extension and direction of axon outgrowth on rigid, but not compliant, substrata. Reducing expression of TRPC1 by antisense morpholinos inhibits the effects of MS channel blockers on axon outgrowth and local Ca(2+) transients. Ca(2+) influx through MS TRPC1 activates the protease calpain, which cleaves the integrin adaptor protein talin to reduce Src-dependent axon outgrowth, likely through altered adhesion turnover. We found that talin accumulates at the tips of dynamic filopodia, which is lost upon cleavage of talin by active calpain. This pathway may also be important in axon guidance decisions since asymmetric inhibition of MS TRPC1 is sufficient to induce growth cone turning. Together our results suggest that Ca(2+) influx through MS TRPC1 on filopodia activates calpain to control growth cone turning during development.
PubMedId: 23283340
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 33, 2013
Authors: Deepika Verma, Nannan Ye, Fanjie Meng, Frederick Sachs, Jason Rahimzadeh, Susan Z Hua
Abstract: Using stress sensitive FRET sensors we have measured cytoskeletal stresses in -actinin and the associated reorganization of the actin cytoskeleton in cells subjected to chronic shear stress. We show that long-term shear stress reduces the average actinin stress and this effect is reversible with removal of flow. The flow-induced changes in cytoskeletal stresses are found to be dynamic, involving a transient decrease in stress (phase-I), a short-term increase (3-6 min) (Phase-II), followed by a longer-term decrease that reaches a minimum in ~20 min (Phase-III), before saturating. These changes are accompanied by reorganization of the actin cytoskeleton from parallel F-actin bundles to peripheral bundles. Blocking mechanosensitive ion channels (MSCs) with Gd(3+) and GsMTx4 (a specific inhibitor) eliminated the changes in cytoskeletal stress and the corresponding actin reorganization, indicating that Ca(2+) permeable MSCs participate in the signaling cascades. This study shows that shear stress induced cell adaptation is mediated via MSCs.
PubMedId: 23028495
Journal-article: PloS one, vol. 7, 2012
Authors: Jinseok Heo, Frederick Sachs, Jianbin Wang, Susan Z Hua
Abstract: Using a microfluidic cell volume sensor we measured the change in the cell volume of Madin-Darby Canine Kidney (MDCK) cells induced by shear stress. An increase in shear stress from 0.2 to 2.0 dyn/cm(2) resulted in a volume decrease to a steady state volume 20 - 30 % smaller than the initial resting cell volume. Independent experiments based on fluorescence quenching confirmed the volume reduction. This shear-induced cell shrinkage was irreversible on the time scale of the experiment ( 30 min). Treatment of 0.1 M Hg(2+) significantly inhibited the volume decrease, suggesting that the shear-induced cell shrinkage is associated with water efflux through aquaporins. The volume decrease cannot be inhibited by 75 mM TEA, 100 M DIDS, or 100 M Gd(3+) suggesting that volume reduction is not directly mediated by K(+) and Cl(-)channels that typically function during regulatory volume decrease (RVD), nor is it through cationic stretch-activated ion channels (SACs). The process also appears to be Ca(2+) independent because it was insensitive to intracellular Ca(2+) level. Since cell volume is determined by the intracellular water content, we postulate that the shear induced reductions in cell volume may arise from increased intracellular hydrostatic pressure as the cell is deformed under flow, which promotes the efflux of water. The increase in internal pressure in a deformable object under the flow is supported by the finite element mechanical model.
PubMedId: 22759987
Journal-article: Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology, vol. 30, 2012
Authors: Philip A Gottlieb, Chilman Bae, Frederick Sachs
Abstract: Piezo1 is a eukaryotic cation-selective mechanosensitive ion channel. To understand channel function in vivo, we first need to analyze and compare the response in the whole cell and the patch. In patches, Piezo1 inactivates and the current is fit well by a 3-state model with a single pressure-dependent rate. However, repeated stimulation led to an irreversible loss of inactivation. Remarkably, the loss of inactivation did not occur on a channel-by-channel basis but on all channels at the same time. Thus, the channels are in common mechanical domain. Divalent ions decreased the unitary conductance from ~68 pS to ~37 pS, irrespective of the cation species. Mg and Ca did not affect inactivation rates, but Zn caused a 3-fold slowing. CytochalasinD (cytoD) does not alter inactivation rates or the transition to the non-inactivating mode but does reduce the steady-state response. Whole-cell currents were similar to patch currents but also had significant differences. In contrast to the patch, cytoD inhibited the current suggesting that the activating forces were transmitted through the actin cytoskeleton. Hypotonic swelling that prestressed the cytoskeleton and the bilayer greatly increased the sensitivity of both control and cytoD cells so there are two pathways to transmit force to the channels. In contrast to patch, removing divalent ions decreased the whole-cell current. The difference between whole cell and patch properties provide new insights into our understanding of the Piezo1 gating mechanisms and cautions against generalization to in situ behavior.
PubMedId: 22790451
Journal-article: Channels (Austin, Tex.), vol. 6, 2012
Authors: Philip A Gottlieb, Frederick Sachs
Abstract: Piezo ion channels have been found to be essential for mechanical responses in cells. These channels were first shown to exist in Neuro2A cells, and the gene was identified by siRNAs that diminished the mechanical response. Piezo channels are approximately 2500 amino acids long, have between 24-32 transmembrane regions, and appear to assemble into tetramers and require no other proteins for activity. They have a reversal potential around 0 mV and show voltage dependent inactivation. The channel is constitutively active in liposomes, indicating that no cytoskeletal elements are required. Heterologous expression of the Piezo protein can create mechanical sensitivity in otherwise insensitive cells. Piezo1 currents in outside-out patches were blocked by the extracellular MSC inhibitor peptide GsMTx4. Both enantiomeric forms of GsMTx4 inhibited channel activity in a manner similar to endogenous mechanical channels. Piezo1 can adopt a tonic (non-inactivating) form with repeated stimulation. The transition to the non-inactivating form generally occurs in large groups of channels, indicating that the channels exist in domains, and once the domain is compromised, the members simultaneously adopt new properties. Piezo proteins are associated with physiological responses in cells, such as the reaction to noxious stimulus of Drosophila larvae. Recent work measuring cell crowding, shows that Piezo1 is essential for the removal of extra cells without apoptosis. Piezo1 mutations have also been linked to the pathological response of red blood cells in a genetic disease called Xerocytosis. These finding suggest that Piezo1 is a key player in cells' responses to mechanical stimuli.
PubMedId: 22790400
Journal-article: Channels (Austin, Tex.), vol. 6, 2012
Authors: Sergei Sukharev, Frederick Sachs
Abstract: Cells perceive force through a variety of molecular sensors, of which the mechanosensitive ion channels are the most efficient and act the fastest. These channels apparently evolved to prevent osmotic lysis of the cell as a result of metabolite accumulation and/or external changes in osmolarity. From this simple beginning, nature developed specific mechanosensitive enzymes that allow us to hear, maintain balance, feel touch and regulate many systemic variables, such as blood pressure. For a channel to be mechanosensitive it needs to respond to mechanical stresses by changing its shape between the closed and open states. In that way, forces within the lipid bilayer or within a protein link can do work on the channel and stabilize its state. Ion channels have the highest turnover rates of all enzymes, and they can act as both sensors and effectors, providing the necessary fluxes to relieve osmotic pressure, shift the membrane potential or initiate chemical signaling. In this Commentary, we focus on the common mechanisms by which mechanical forces and the local environment can regulate membrane protein structure, and more specifically, mechanosensitive ion channels.
PubMedId: 22797911
Journal-article: Journal of cell science, vol. 125, 2012
Authors: Fanjie Meng, Frederick Sachs
Abstract: Mechanical stress is an unmapped source of free energy in cells. Mapping the stress fields in a heterogeneous time-dependent environment like that found in cells requires probes that are specific for different proteins and respond to biologically relevant forces with minimal disturbance to the host system. To meet these goals, we have designed a genetically encoded stress sensor with minimal volume and high sensitivity and dynamic range. The new FRET-based sensor, called cpstFRET, is designed to be modulated by the angles between the donor and acceptor rather than the distance between them. Relative to other probes, it is physically smaller and exhibits a greater dynamic range and sensitivity and expresses well. For in vivo testing, we measured stress gradients in time and space in non-erythroid spectrin in several different cell types and found that spectrin is under constitutive stress in some cells but not in others. Stresses appear to be generated by both F-actin and tubulin. The probe revealed, for the first time, that spectrin undergoes time-dependent force modulation during cell migration. cpstFRET can be employed in vitro, in vivo and in situ, and when incorporated into biologically expressed extracellular polymers such as collagen, it can report multidimensional stress fields.
PubMedId: 22389408
Journal-article: Journal of cell science, vol. 125, 2012
Authors: R??mi Peyronnet, Reza Sharif-Naeini, Joost??H.A. Folgering, Malika Arhatte, Martine Jodar, Charbel El??Boustany, Claire Gallian, Michel Tauc, Christophe Duranton, Isabelle Rubera, Florian Lesage, York Pei, Dorien??J.M. Peters, Stefan Somlo, Frederick Sachs, Amanda Patel, Eric Honor??, Fabrice Duprat
PubMedId: 22832196
Journal-article: Cell Reports, vol. 1, pp. 241-250, 2012
Authors: Gregory B Kowalsky, Derek Beam, Myung J Oh, Frederick Sachs, Susan Z Hua, Irena Levitan
Abstract: The maintenance of cell volume homeostasis is critical for preventing pathological cell swelling that may lead to severe cellular dysfunction or cell death. Our earlier studies have shown that volume-regulated anion channels that play a major role in the regulation of cell volume are facilitated by a decrease in cellular cholesterol suggesting that cholesterol depletion should also facilitate regulatory volume decrease (RVD), the ability of cells to recover from hypotonic swelling. In this study, we test this hypothesis using a novel methodology developed to measure changes in cell volume using a microfluidics chamber. Our data show that cholesterol depletion of Chinese Hamster Ovary (CHO) significantly facilitates the recovery process, as is apparent from a faster onset of the RVD (16210 s. vs. 1145 s. in control and cholesterol depleted cells respectively) and a higher degree of volume recovery after 10 min of the hypotonic challenge (41%6% vs. 65%6% in control and cholesterol depleted cells respectively). In contrast, enriching cells with cholesterol had no effect on the RVD process. We also show here that similarly to our previous observations in endothelial cells, cholesterol depletion significantly increases the stiffness of CHO cells suggesting that facilitation of RVD may be associated with cell stiffening. Furthermore, we also show that increasing cell stiffness by stabilizing F-actin with jasplakinolide also facilitates RVD development. We propose that cell stiffening enhances cell mechano-sensitivity, which in turn facilitates the RVD process.
PubMedId: 22179012
Journal-article: Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology, vol. 28, 2011
Authors: Jason Rahimzadeh, Fanjie Meng, Fredrick Sachs, Jianbin Wang, Deepika Verma, Susan Z Hua
Abstract: The mechanical stress due to shear flow has profound effects on cell proliferation, transport, gene expression, and apoptosis. The mechanisms for flow sensing and transduction are unclear, but it is postulated that fluid flow pulls upon the apical surface, and the resulting stress is eventually transmitted through the cytoskeleton to adhesion plaques on the basal surface. Here we report a direct observation of this flow-induced stress in the cytoskeleton in living cells using a parallel plate microfluidic chip with a fluorescence resonance energy transfer (FRET)-based mechanical stress sensor in actinin. The sensing cassette was genetically inserted into the cytoskeletal host protein and transfected into Madin-Darby canine kidney cells. A shear stress of 10 dyn/cm(2) resulted in a rapid increase in the FRET ratio indicating a decrease in stress across actinin with flow. The effect was reversible, and cells were able to respond to repeated stimulation and showed adaptive changes in the cytoskeleton. Flow-induced Ca(2+) elevation did not affect the response, suggesting that flow-induced changes in actinin stress are insensitive to intracellular Ca(2+) level. The reduction in FRET ratio suggests actin filaments are under normal compression in the presence of flow shear stress due to changes in cell shape, and/or actinin is not in series with actin. Treatment with cytochalasin-D that disrupts F-actin reduced prestress and the response to flow. The FRET/flow method is capable of resolving changes of stress in multiple proteins with optical spatial resolution and time resolution >1 Hz. This promises to provide insight into the force distribution and transduction in all cells.
PubMedId: 21653900
Journal-article: American journal of physiology. Cell physiology, vol. 301, 2011
Authors: Chilman Bae, Frederick Sachs, Philip A Gottlieb
Abstract: Cells can respond to mechanical stress by gating mechanosensitive ion channels (MSCs). The cloning of Piezo1, a eukaryotic cation selective MSC, defines a new system for studying mechanical transduction at the cellular level. Because Piezo1 has electrophysiological properties similar to those of endogenous cationic MSCs that are selectively inhibited by the peptide GsMTx4, we tested whether the peptide targets Piezo1 activity. Extracellular GsMTx4 at micromolar concentrations reversibly inhibited 80% of the mechanically induced current of outside-out patches from transfected HEK293 cells. The inhibition was voltage insensitive, and as seen with endogenous MSCs, the mirror image d enantiomer inhibited like the l. The rate constants for binding and unbinding based on Piezo1 current kinetics provided association and dissociation rates of 7.0 10(5) M(-1) s(-1) and 0.11 s(-1), respectively, and a K(D) of 155 nM, similar to values previously reported for endogenous MSCs. Consistent with predicted gating modifier behavior, GsMTx4 produced an 30 mmHg rightward shift in the pressure-gating curve and was active on closed channels. In contrast, streptomycin, a nonspecific inhibitor of cationic MSCs, showed the use-dependent inhibition characteristic of open channel block. The peptide did not block currents of the mechanical channel TREK-1 on outside-out patches. Whole-cell Piezo1 currents were also reversibly inhibited by GsMTx4, and although the off rate was nearly identical to that of outside-out patches, differences were observed for the on rate. The ability of GsMTx4 to target the mechanosensitivity of Piezo1 supports the use of this channel in high-throughput screens for pharmacological agents and diagnostic assays.
PubMedId: 21696149
Journal-article: Biochemistry, vol. 50, 2011
Authors: Yuexiu Wang, Fanjie Meng, Frederick Sachs
Keywords: biomechanics cytoskeleton fluorescence resonance energy transfer force sensor mechanical force stress
Abstract: THERE ARE THREE SOURCES OF FREE ENERGY FOR CELLS: chemical potential, electrical potential and mechanical potential. There is little known about the last one since there have not been simple ways to measure stress in proteins in cells. we have now developed genetically encoded force sensors to assess the stress in fibrous proteins in living cells. These FReT based fluorescence sensors can be read out at video rates and provide real time maps of the stress distribution in cells, tissues and animals. The sensors can be inserted into specific proteins and in general do not disturb the normal function or anatomy. The original sensors used mutant GFPs linked by elastic linkers. These sensors provide a linear output with applied stress but the response is linear in strain. To improve contrast and dynamic range we have now developed a new class of sensors that are smaller making them less invasive, and have much higher intrinsic sensitivity since force modulates the angle between the donor and acceptor much more than the distance between them. Known as cpstFRET, the probe shows improved biocompatibility, wider dynamic range and higher sensitivity.
PubMedId: 21966553
Journal-article: Communicative & integrative biology, vol. 4, 2011
Authors: Ce??dric Bathany, Derek Beahm, James D. Felske, Frederick Sachs, Susan Z. Hua
Abstract: This paper describes a microfluidic-based assay capable of measuring gap-junction mediated dye diffusion in cultured cells. The technique exploits multistream laminar flow to selectively expose cells to different environments, enabling continuous loading of cells in one compartment while monitoring, in real time, dye diffusion into cells of a neighboring compartment. A simple one-dimensional diffusion model fit to the data extracted the diffusion coefficient of four different dyes, 5-(6)-carboxyfluorescein, 5-chloromethylfluorescein, Oregon green 488 carboxylic acid, and calcein. Different inhibitors were assayed for their ability to reduce dye coupling. The chip can screen multiple inhibitors in parallel in the same cell preparation, demonstrating its potential for high throughput. The technique provides a convenient method to measure gap junction mediated diffusion and a screen for drugs that affect gap junction communication.
PubMedId: 21182279
Journal-article: Analytical Chemistry, vol. 83, pp. 933-939, 2011
Authors: T.A. Quinn, S. Granite, M.A. Allessie, C. Antzelevitch, C. Bollensdorff, G. Bub, R.A.B. Burton, E. Cerbai, P.S. Chen, M. Delmar, D. DiFrancesco, Y.E. Earm, I.R. Efimov, M. Egger, E. Entcheva, M. Fink, R. Fischmeister, M.R. Franz, A. Garny, W.R. Giles, T. Hannes, S.E. Harding, P.J. Hunter, G. Iribe, J. Jalife, C.R. Johnson, R.S. Kass, I. Kodama, G. Koren, P. Lord, V.S. Markhasin, S. Matsuoka, A.D. McCulloch, G.R. Mirams, G.E. Morley, S. Nattel, D. Noble, S.P. Olesen, A.V. Panfilov, N.A. Trayanova, U. Ravens, S. Richard, D.S. Rosenbaum, Y. Rudy, F. Sachs, F.B. Sachse, D.A. Saint, U. Schotten, O. Solovyova, P. Taggart, L. Tung, A. Varr??, P.G. Volders, K. Wang, J.N. Weiss, E. Wettwer, E. White, R. Wilders, R.L. Winslow, P. Kohl
PubMedId: 21745496
Journal-article: Progress in Biophysics and Molecular Biology, vol. 107, pp. 4-10, 2011
Authors: Frederick Sachs
Book-chapter: Cardiac Mechano-Electric Coupling and Arrhythmias, pp. 11-18, 2011
Authors: Fanjie Meng, Frederick Sachs
Abstract: Mechanical forces are ubiquitous modulators of cell activity but little is known about the mechanical stresses in the cell. Genetically encoded FRET-based force sensors now allow the measurement of local stress in specific host proteins in vivo in real time. For a minimally invasive probe, we designed one with a mechanical compliance matching that of many common cytoskeleton proteins. sstFRET is a cassette composed of Venus and Cerulean linked by a spectrin repeat. The stress sensitivity of the probe was measured in solution using DNA springs to push the donor and acceptor apart with 5-7 pN and this produced large changes in FRET. To measure cytoskeletal stress in vivo we inserted sstFRET into -actinin and expressed it in HEK and BAEC cells. Time-lapse imaging showed the presence of stress gradients in time and space, often uncorrelated with obvious changes in cell shape. The gradients could be rapidly relaxed by thrombin-induced cell contraction associated with inhibition of myosin II. The tension in actinin fluctuated rapidly (scale of seconds) illustrating a cytoskeleton in dynamic equilibrium. Stress in the cytoskeleton can be driven by macroscopic stresses applied to the cell. Using sstFRET as a tool to measure internal stress, we tested the prediction that osmotic pressure increases cytoskeletal stress. As predicted, hypotonic swelling increased the tension in actinin, confirming the model derived from AFM. Anisotonic stress also produced a novel transient (~2 minutes) decrease in stress upon exposure to a hypotonic challenge, matched by a transient increase with hypertonic stress. This suggests that, at rest, the stress axis of actinin is not parallel to the stress axis of actin and that swelling can reorient actinin to lie more parallel where it can absorb a larger fraction of the total stress. Protein stress sensors are opening new perspectives in cell biology.
PubMedId: 21172803
Journal-article: Journal of cell science, vol. 124, 2010
Authors: Arthur Beyder, James L Rae, Cheryl Bernard, Peter R Strege, Frederick Sachs, Gianrico Farrugia
Abstract: The voltage-sensitive sodium channel Na(v)1.5 (encoded by SCN5A) is expressed in electromechanical organs and is mechanosensitive. This study aimed to determine the mechanosensitive transitions of Na(v)1.5 at the molecular level. Na(v)1.5 was expressed in HEK 293 cells and mechanosensitivity was studied in cell-attached patches. Patch pressure up to -50 mmHg produced increases in current and large hyperpolarizing shifts of voltage dependence with graded shifts of half-activation and half-inactivation voltages (V(1/2)) by 0.7 mV mmHg(-1). Voltage dependence shifts affected channel kinetics by a single constant. This suggested that stretch accelerated only one of the activation transitions. Stretch accelerated voltage sensor movement, but not rate constants for gate opening and fast inactivation. Stretch also appeared to stabilize the inactivated states, since recovery from inactivation was slowed with stretch. Unitary conductance and maximum open probability were unaffected by stretch, but peak current was increased due to an increased number of active channels. Stretch effects were partially reversible, but recovery following a single stretch cycle required minutes. These data suggest that mechanical activation of Na(v)1.5 results in dose-dependent voltage dependence shifts of activation and inactivation due to mechanical modulation of the voltage sensors.
PubMedId: 21041530
Journal-article: The Journal of physiology, vol. 588, 2010
Authors: Kishore Kamaraju, Philip A Gottlieb, Frederick Sachs, Sergei Sukharev
Abstract: GsMTx4 is a 34-residue peptide isolated from the tarantula Grammostola spatulata folded into an inhibitory cysteine knot and it selectively affects gating of some mechanosensitive channels. Here we report the effects of cytoplasmic GsMTx4 on the two bacterial channels, MscS and MscL, in giant Escherichia coli spheroplasts. In excised inside-out patches, GsMTx4 sensitized both channels to tension by increasing the opening rate and decreasing the closing rate. With ascending and descending pressure ramps, GsMTx4 increased the gating hysteresis for MscS, a consequence of slower gating kinetics. Quantitative kinetic analysis of the primary CO transition showed that the hysteresis is a result of the decreased closing rate. The gating barrier location relative to the open state energy well was unaffected by GsMTx4. A reconstructed energy profile suggests that the peptide prestresses the resting state of MscS, lowering the net barrier to opening and stabilizes the open conformation by 8 kT. In excised patches, both MscL and MscS exhibit reversible adaptation, a process separable from inactivation for MscS. GsMTx4 decreased the rate of reversible adaptation for both channels and the MscS recovery rate from the inactivation. These measurements support a mechanism where GsMTx4 binds to the lipid interface of the channel, increasing the local stress that is sensed by the channels and stabilizing the expanded conformations.
PubMedId: 21044584
Journal-article: Biophysical journal, vol. 99, 2010
Authors: Philip A Gottlieb, Tara Barone, Frederick Sachs, Robert Plunkett
Abstract: GsMTx4, a peptide inhibitor for mechanosensitive ion channels (MSCs), promoted neurite outgrowth from PC12 cells in the presence of NGF in a dose-dependent manner between 5 and 100 microM peptide. Enhanced neurite growth required >12 h of peptide exposure in cells grown with NGF. Adsorption of GsMTx4 to serum proteins in the media lowered the free peptide concentration of 100 microM to a free concentration of 5 microM, a concentration shown to completely inhibit MSCs in the patch clamp assay. Outside-out patches from PC12 cells grown in NGF had mechanically activated cation channels that were reversibly inhibited by GsMTx4. These results are similar to those observed by Gomez and co-workers in Xenopus spinal cord. The inhibition of mechanosensitive channels by GsMTx4 may be a useful approach to accelerate regeneration of neurons in neurodegenerative diseases and spinal cord injury.
PubMedId: 20600595
Journal-article: Neuroscience letters, vol. 481, 2010
Authors: Susan Z Hua, Philip A Gottlieb, Jinseok Heo, Frederick Sachs
Abstract: Cells respond to a hyposmotic challenge by swelling and then returning toward the resting volume, a process known as the regulatory volume decrease or RVD. The sensors for this process have been proposed to include cationic mechanosensitive ion channels that are opened by membrane tension. We tested this hypothesis using a microfluidic device to measure cell volume and the peptide GsMTx4, a specific inhibitor of cationic mechanosensitive channels. GsMTx4 had no effect on RVD in primary rat astrocytes or Madin-Darby canine kidney (MDCK) cells but was able to completely inhibit RVD and the associated Ca(2+) uptake in normal rat kidney (NRK-49F) cells in a dose-dependent manner. Gadolinium (Gd(3+)), a nonspecific blocker of many mechanosensitive channels, inhibited RVD and Ca(2+) uptake in all three cell types, demonstrating the existence of at least two types of volume sensors. Single-channel stretch-activated currents are present in outside-out patches from NRK-49F, MDCK, and astrocytes, and they are reversibly inhibited by GsMTx4. While mechanosensitive channels are involved in volume regulation, their role for volume sensing is specialized. The NRK cells form a stable platform from which to screen drugs that affect volume regulation via mechanosensory channels and as a sensitive system to clone the channel.
PubMedId: 20457830
Journal-article: American journal of physiology. Cell physiology, vol. 298, 2010
Authors: Frederick Sachs
Abstract: Mechanosensitive ion channels (MSCs) exist in all cells, but mechanosensitivity is a phenotype not a genotype. Specialized mechanoreceptors such as the hair cells of the cochlea require elaborate mechanical impedance matching to couple the channels to the external stress. In contrast, MSCs in nonspecialized cells appear activated by stress in the bilayer local to the channel--within about three lipids. Local mechanical stress can be produced by far-field tension, amphipaths, phase separations, the cytoskeleton, the extracellular matrix, and the adhesion energy between the membrane and a patch pipette. Understanding MSC function requires under standing the stimulus.
PubMedId: 20134028
Journal-article: Physiology (Bethesda, Md.), vol. 25, 2010
Authors: F Sachs, W E Brownell, A G Petrov
PubMedId: 20165559
Journal Article: MRS bulletin, vol. 34, 2010
Authors: David H Vandorpe, Chang Xu, Boris E Shmukler, Leo E Otterbein, Marie Trudel, Frederick Sachs, Philip A Gottlieb, Carlo Brugnara, Seth L Alper
Abstract: Deoxygenation of sickle erythrocytes activates a cation permeability of unknown molecular identity (Psickle), leading to elevated intracellular [Ca(2+)] ([Ca(2+)](i)) and subsequent activation of K(Ca) 3.1. The resulting erythrocyte volume decrease elevates intracellular hemoglobin S (HbSS) concentration, accelerates deoxygenation-induced HbSS polymerization, and increases the likelihood of cell sickling. Deoxygenation-induced currents sharing some properties of Psickle have been recorded from sickle erythrocytes in whole cell configuration. We now show by cell-attached and nystatin-permeabilized patch clamp recording from sickle erythrocytes of mouse and human that deoxygenation reversibly activates a Ca(2+)- and cation-permeable conductance sensitive to inhibition by Grammastola spatulata mechanotoxin-4 (GsMTx-4; 1 microM), dipyridamole (100 microM), DIDS (100 microM), and carbon monoxide (25 ppm pretreatment). Deoxygenation also elevates sickle erythrocyte [Ca(2+)](i), in a manner similarly inhibited by GsMTx-4 and by carbon monoxide. Normal human and mouse erythrocytes do not exhibit these responses to deoxygenation. Deoxygenation-induced elevation of [Ca(2+)](i) in mouse sickle erythrocytes did not require KCa3.1 activity. The electrophysiological and fluorimetric data provide compelling evidence in sickle erythrocytes of mouse and human for a deoxygenation-induced, reversible, Ca(2+)-permeable cation conductance blocked by inhibition of HbSS polymerization and by an inhibitor of strctch-activated cation channels. This cation permeability pathway is likely an important source of intracellular Ca(2+) for pathologic activation of KCa3.1 in sickle erythrocytes. Blockade of this pathway represents a novel therapeutic approach for treatment of sickle disease.
PubMedId: 20090940
Journal-article: PloS one, vol. 5, 2009
Authors: Haixia Huang, Hua Wei, Ping Liu, Wei Wang, Frederick Sachs, Weizhen Niu
Abstract: Transient stretching of the ventricle can trigger arrhythmias and evoke ventricular fibrillation, especially when the stimulation occurs in the vulnerable period. To explore the sensitivity of small hearts we used a commercial pressure servo to study the kinetic relationship of left ventricular pressure to excitability and arrhythmias in the rat heart. Stimulation protocols were readily composed on the computer and programmed to vary the stimulus amplitude and timing relative to pacing. The pressure-induced premature ventricular excitations were similar to those observed in larger hearts, but the convenience of using small hearts allows the use of inexpensive transgenic animals to explore the molecular basis of transduction.
PubMedId: 19592413
Journal-article: Experimental physiology, vol. 94, 2009
Authors: Thomas M Suchyna, Vladislav S Markin, Frederick Sachs
Abstract: Interpreting channel behavior in patches requires an understanding of patch structure and dynamics, especially in studies of mechanosensitive channels. High resolution optical studies show that patch formation occurs via blebbing that disrupts normal membrane structure and redistributes in situ components including ion channels. There is a 1-2 microm region of the seal below the patch where proteins are excluded and this may consist of extracted lipids that form the gigaseal. Patch domes often have complex geometries with inhomogeneous stresses due to the membrane-glass adhesion energy (E(a)), cytoskeletal forces, and possible lipid subdomains. The resting tension in the patch dome ranges from 1-4 mN/m, a significant fraction of the lytic tension of a bilayer ( approximately 10 mN/m). Thus, all patch experiments are conducted under substantial, and uneven, resting tension that may alter the kinetics of many channels. E(a) seems dominated by van der Waals attraction overlaid with a normally repulsive Coulombic force. High ionic strength pipette saline increased E(a) and, surprisingly, increased cytoskeletal rigidity in cell-attached patches. Low pH pipette saline also increased E(a) and reduced the seal selectivity for cations, presumably by neutralizing the membrane surface charge. The seal is a negatively charged, cation selective, space with a resistance of approximately 7 gigohm/microm in 100 mM KCl, and the high resistivity of the space may result from the presence of high viscosity glycoproteins. Patches creep up the pipette over time with voltage independent and voltage dependent components. Voltage-independent creep is expected from the capillary attraction of E(a) and the flow of fresh lipids from the cell. Voltage-dependent creep seems to arise from electroosmosis in the seal. Neutralization of negative charges on the seal membrane with low pH decreased the creep rate and reversed the direction of creep at positive pipette potentials.
PubMedId: 19651032
Journal-article: Biophysical journal, vol. 97, 2009
Authors: Arthur Beyder, Frederick Sachs
Abstract: Membranes flex with changes in transmembrane potential as a result of changes in interfacial tension, the Lippman effect. We studied the membrane electromotility of Shaker K(+)-transfected HEK-293 cells in real time by using combined patch-clamp atomic force microscopy. In the voltage range where the channels were closed, Shaker expression had little effect on electromotility relative to wild-type cells. Depolarization between -120 and -40 mV resulted in a linear upward cantilever deflection equivalent to an increase in membrane tension. However, when depolarized sufficiently for channel opening, the electromotility saturated and only recovered over 10 s of milliseconds. This remarkable loss of motility was associated with channel opening, not ionic flux or movement of the voltage sensors. The IL mutant of Shaker, in which the voltage dependence of channel opening but not sensor movement is shifted to more positive potentials, caused the loss of electromotility saturation also to shift to more positive potentials. The temporary loss of electromotility associated with channel opening is probably caused by local buckling of the bilayer as the inner half of the channel expands as expected from X-ray structural data.
PubMedId: 19366664
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 106, 2009
Authors: Reza Sharif-Naeini, Joost H.A. Folgering, Delphine Bichet, Fabrice Duprat, Inger Lauritzen, Malika Arhatte, Martine Jodar, Alexandra Dedman, Franck C. Chatelain, Uwe Schulte, Kevin Retailleau, Laurent Loufrani, Amanda Patel, Frederick Sachs, Patrick Delmas, Dorien J.M. Peters, Eric Honor??
PubMedId: 19879844
Journal-article: Cell, vol. 139, pp. 587-596, 2009
Authors: F. Sachs, W. E. Brownell, A. G. Petrov
Journal-article: MRS Bulletin, vol. 34, pp. 665-670, 2009
Authors: Xuxia Liu, Haixia Huang, Wei Wang, Jun Wang, Frederick Sachs, Weizhen Niu
Abstract: Stress in the lipids of the cell membrane may be responsible for activating stretch-activated channels (SACs) in nonspecialized sensory cells such as cardiac myocytes, where they are likely to play a role in cardiac mechanoelectric feedback. We examined the influence of the mechanical microenvironment on the gating of stretch-activated potassium channels (SAKCs) in rat atrial myocytes. The goal was to examine the role of the cytoskeleton in the gating process. We recorded from blebs that have minimal cytoskeleton and cells treated with cytochalasin B (cyto-B) to disrupt filamentous actin. Histochemical and electron microscopic techniques confirmed that the bleb membrane was largely free of F-actin. Channel currents showed mechanosensitivity and potassium selectivity and were activated by low pH and arachidonic acid, similar to properties of TREK-1. Some patches showed a time-dependent decrease in current that may be adaptation or inactivation, and since this decrease appeared in control cells and blebs, it is probably not the result of adaptation in the cytoskeleton. Cyto-B treatment and blebbing caused an increase in background channel activity, suggesting a transfer of stress from actin to bilayer and then to the channel. The slope sensitivity of gating before and after cyto-B treatment was similar to that of blebs, implying the characteristic change of dimensions associated with channel gating was the same in the three mechanical environments. The mechanosensitivity of SAKCs appears to be the result of interaction with membrane lipids and not of direct involvement of the cytoskeleton.
PubMedId: 19015804
Journal-article: The Journal of membrane biology, vol. 226, 2008
Authors: Chiara Spagnoli, Arthur Beyder, Stephen Besch, Frederick Sachs
Abstract: Cells swell in response a hypoosmotic challenge. By converting osmotic pressure to hydrostatic pressure at the cell membrane via van't Hoff's law, and converting that to tension via Laplace's law one predicts that the cell membrane should stretch and become stiff. We tested this prediction using the atomic force microscopy. During osmotic swelling cells did not become stiff and generally became softer. This result contradicts the assumption of the cell membrane as the constraining element in osmotic stress but is consistent with the cytoskeleton acting as a cross-linked gel. Models of the cells' response to osmotic stress must include energy terms for three-dimensional stresses.
PubMedId: 18851074
Journal-article: Physical review. E, Statistical, nonlinear, and soft matter physics, vol. 78, 2008
Authors: Fanjie Meng, Thomas M Suchyna, Frederick Sachs
Abstract: To measure mechanical stress in real time, we designed a fluorescence resonance energy transfer (FRET) cassette, denoted stFRET, which could be inserted into structural protein hosts. The probe was composed of a green fluorescence protein pair, Cerulean and Venus, linked with a stable alpha-helix. We measured the FRET efficiency of the free cassette protein as a function of the length of the linker, the angles of the fluorophores, temperature and urea denaturation, and protease treatment. The linking helix was stable to 80 degrees C, unfolded in 8 m urea, and rapidly digested by proteases, but in all cases the fluorophores were unaffected. We modified the alpha-helix linker by adding and subtracting residues to vary the angles and distance between the donor and acceptor, and assuming that the cassette was a rigid body, we calculated its geometry. We tested the strain sensitivity of stFRET by linking both ends to a rubber sheet subjected to equibiaxial stretch. FRET decreased proportionally to the substrate strain. The naked cassette expressed well in human embryonic kidney-293 cells and, surprisingly, was concentrated in the nucleus. However, when the cassette was located into host proteins such alpha-actinin, nonerythrocyte spectrin and filamin A, the labeled hosts expressed well and distributed normally in cell lines such as 3T3, where they were stressed at the leading edge of migrating cells and relaxed at the trailing edge. When collagen-19 was labeled near its middle with stFRET, it expressed well in Caenorhabditis elegans, distributing similarly to hosts labeled with a terminal green fluorescent protein, and the worms behaved normally.
PubMedId: 18479457
Journal-article: The FEBS journal, vol. 275, 2008
Authors: Thomas Pennell, Thomas Suchyna, Jianbin Wang, Jinseok Heo, James D Felske, Frederick Sachs, Susan Z Hua
Abstract: We developed a microfluidic chip that provides rapid temperature changes and accurate temperature control of the perfusing solution to facilitate patch-clamp studies. The device consists of a fluid channel connected to an accessible reservoir for cell culture and patch-clamp measurements. A thin-film platinum heater was placed in the flow channel to generate rapid temperature change, and the temperature was monitored using a thin-film resistor. We constructed the thermal chip using SU-8 on a glass wafer to minimize the heat loss. The chip is capable of increasing the solution temperature from bath temperature (20 degrees C) to 80 degrees C at an optimum heating rate of 0.5 degrees C/ms. To demonstrate the ability of the thermal chip, we have conducted on-chip patch-clamp recordings of temperature-sensitive ion channels (TRPV1) transfected HEK293 cells. The heat-stimulated currents were observed using whole-cell and cell-attached patch configurations. The results demonstrated that the chip can provide rapid temperature jumps at the resolution of single-ion channels.
PubMedId: 18302344
Journal-article: Analytical chemistry, vol. 80, 2008
Authors: J. Kockskamper, M. Khafaga, M. Grimm, A. Elgner, S. Walther, A. Kockskamper, D. von Lewinski, H. Post, M. Grossmann, H. Dorge, P. A. Gottlieb, F. Sachs, T. Eschenhagen, F. A. Schondube, B. Pieske
PubMedId: 18503051
Journal-article: Cardiovascular Research, vol. 79, pp. 642-651, 2008
Authors: Jens Kocksk??mper, Dirk von Lewinski, Mounir Khafaga, Andreas Elgner, Michael Grimm, Thomas Eschenhagen, Philip A. Gottlieb, Frederick Sachs, Burkert Pieske
PubMedId: 18466959
Journal-article: Progress in Biophysics and Molecular Biology, vol. 97, pp. 250-267, 2008
Authors: Jinseok Heo, Fanjie Meng, Frederick Sachs, Susan Z Hua
Abstract: Using a microfluidic volume sensor, we studied the dynamic effects of Hg2+ on hypotonic stress-induced volume changes in CHO cells. A hypotonic challenge to control cells caused them to swell but did not evoke a significant regulatory volume decrease (RVD). Treatment with 100 muM HgCl2 caused a substantial increase in the steady-state volume following osmotic stress. Continuous hypotonic challenge following a single 10-min exposure to HgCl2 produced a biphasic volume increase with a steady-state volume 100% larger than control cells. Repeated hypotonic challenges to cells exposed once to Hg2+ resulted in a sequential approach to the same steady-state volume. Stimulation after reaching steady state caused a reduction in peak cell volume. Repeated stimulation was different than continuous stimulation resulting in a more rapid approach to steady state. Substituting extracellular Na+ with impermeant NMDG+ in the hypotonic solution produced a rapid RVD-like volume decrease and eliminated the Hg2+-induced excess swelling. The volume decrease in the presence of Hg2+ was inhibited by tetraethylammonium and 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid disodium, blockers of K+ and Cl(-) channels, respectively, suggesting that part of the Hg2+ effect was increasing NaCl influx over KCl efflux. The presence of multiple phases of steady-state volume and their sensitivity to the stimulation history suggests that factors beyond solute fluxes, such as modification of mechanical stress within the cytoskeleton also plays a role in the response to hypotonic stress.
PubMedId: 18365146
Journal-article: Cell biochemistry and biophysics, vol. 51, 2007
Authors: Thomas M Suchyna, Frederick Sachs
Abstract: Muscular dystrophy is associated with increased activity of mechanosensitive channels (MSCs) and increased cell calcium levels. MSCs in patches from mdx mouse myotubes have higher levels of resting activity, compared to patches from wild-type mice, and a pronounced latency of activation and deactivation. Measurements of patch capacitance and geometry reveal that the differences are linked to cortical membrane mechanics rather than to differences in channel gating. We found unexpectedly that patches from mdx mice are strongly curved towards the pipette tip by actin pulling normal to the membrane. This force produces a substantial tension (approximately 5 mN m(-1)) that can activate MSCs in the absence of overt stimulation. The inward curvature of patches from mdx mice is eliminated by actin inhibitors. Applying moderate suction to the pipette flattens the membrane, reducing tension, and making the response appear to be stretch inactivated. The pronounced latency to activation in patches from mdx mice is caused by the mechanical relaxation time required to reorganize the cortex from inward to outward curvature. The increased latency is equivalent to a three-fold increase in cortical viscosity. Disruption of the cytoskeleton by chemical or mechanical means eliminates the differences in kinetics and curvature between patches from wild-type and mdx mice. The stretch-induced increase in specific capacitance of the patch, approximately 80 fF microm(-2), far exceeds the specific capacitance of bilayers, suggesting the presence of stress-sensitive access to large pools of membrane, possibly caveoli, T-tubules or portions of the gigaseal. In mdx mouse cells the intrinsic gating property of fast voltage-sensitive inactivation is lost. It is robust in wild-type mouse cells (observed in 50% of outside-out patches), but never observed in mdx cells. This link between dystrophin and inactivation may lead to increased background cation currents and Ca2+ influx. Spontaneous Ca2+ transients in mdx mouse cells are sensitive to depolarization and are inhibited by the specific MSC inhibitor GsMTx4, in both the D and L forms.
PubMedId: 17255168
Journal-article: The Journal of physiology, vol. 581, 2007
Authors: Chiara Spagnoli, Arthur Beyder, Stephen R Besch, Frederick Sachs
Abstract: Atomic force microscopy (AFM) is used to study the morphological and mechanical properties of living cells. However, experiments performed over minutes to hours are subject to significant instrumental drift. The main sources of drift are the cantilever's geometrical asymmetry and bimorphic construction. We developed a simple software Stick-and-Move (SaM) routine for AFM that eliminates drift by continuously referencing the sample position to the substrate while acquiring force-distance curves. Control experiments show no drift over 15 min at an acquisition rate of 0.1 Hz. As a proof of concept, we applied the SaM to study the response of rat astrocytes to osmotic stress, observing dimensional and constitutive changes during volume regulation.
PubMedId: 17411236
Journal-article: The Review of scientific instruments, vol. 78, 2007
Authors: Philip A. Gottlieb, Thomas M. Suchyna, Frederick Sachs
PubMedId: 25168134
Book-chapter: Mechanosensitive Ion Channels, Part B, pp. 81-109, 2007
Authors: Philip Gottlieb, Joost Folgering, Rosario Maroto, Albert Raso, Thomas G. Wood, Alex Kurosky, Charles Bowman, Delphine Bichet, Amanda Patel, Frederick Sachs, Boris Martinac, Owen P. Hamill, Eric Honor??
PubMedId: 17957383
Journal-article: Pfl??gers Archiv - European Journal of Physiology, vol. 455, pp. 1097-1103, 2007
Authors: Jean Chemin, Amanda Jane Patel, Fabrice Duprat, Frederick Sachs, Michel Lazdunski, Eric Honore
Abstract: TREK-1 is an unconventional K(+) channel that is activated by both physical and chemical stimuli. In this study, we show that the inner leaflet membrane phospholipids, including PIP(2), exert a mixed stimulatory and inhibitory effect on TREK-1. Intra-cellular phospholipids inhibit basal channel activity and activation by membrane stretch, intra-cellular acidosis and arachidonic acid. However, binding of endogenous negative inner leaflet phospholipids with poly-lysine reduces inhibition and reveals channel stimulation by exogenous intra-cellular phospholipids. A similar effect is observed with PI, PE, PS and PA, unlike DG, demonstrating that the phosphate at position 3 is required although the global charge of the molecule is not critical. Inhibition depends on the distal C-terminal domain that conditions channel mechano-sensitivity, but is independent of the positively charged PIP(2) stimulatory site in the proximal C-terminal domain. This is, to our knowledge, the first report of an ion channel dually regulated by membrane phospholipids.
PubMedId: 17384962
Journal-article: Pflugers Archiv : European journal of physiology, vol. 455, 2006
Authors: Charles L Bowman, Philip A Gottlieb, Thomas M Suchyna, Yolanda K Murphy, Frederick Sachs
Abstract: Sensing the energy from mechanical inputs is ubiquitous--and perhaps the oldest form of biological energy transduction. However, the tools available to probe the mechanisms of transduction are far fewer than for the chemical and electric field sensitive transducers. The one pharmacological tool available for mechansensitive ion channels (MSCs) is a peptide (GsMTx-4) isolated from venom of the tarantula, Grammostola spatulata, that blocks cationic MSCs found in non-specialized eukaryotic tissues. In this review, we summarize the current knowledge of GsMTx-4, and discuss the inevitable crosstalk between the MSC behavior and the mechanical properties of the cell cortex.
PubMedId: 17157345
Journal-article: Toxicon : official journal of the International Society on Toxinology, vol. 49, 2006
Authors: Nicholas P Whitehead, Margaret Streamer, Lynn I Lusambili, Frederick Sachs, David G Allen
Abstract: It is well-known that muscles from mdx mice are more susceptible to membrane damage from eccentric contractions than wild-type muscles. The present study tested the hypothesis that the stretch-induced membrane permeability in dystrophic muscle is due to Ca(2+) entry through stretch-activated channels (SACs) and the subsequent activation of Ca(2+) -dependent degradative pathways. Eccentric contractions were carried out on muscles from mdx and wild-type mice, both on isolated muscles and on intact mice subjected to downhill running on a treadmill. In isolated muscles the SAC blockers, streptomycin and GsMTx4, improved force and significantly reduced the uptake of procion orange dye into fibres from mdx muscles, which increased progressively over 60 min after the eccentric contractions. In experiments on intact mdx mice, streptomycin also partially prevented the reduced force and the increased membrane permeability (Evans Blue Dye uptake). The results suggest that Ca(2+) entry through SACs activates Ca(2+) -dependent pathways, which are the main cause of the increased membrane permeability in mdx muscle.
PubMedId: 17005404
Journal-article: Neuromuscular disorders : NMD, vol. 16, 2006
Authors: Lorin S Milescu, Ahmet Yildiz, Paul R Selvin, Frederick Sachs
Abstract: Processive molecular motors, such as kinesin, myosin, or dynein, convert chemical energy into mechanical energy by hydrolyzing ATP. The mechanical energy is used for moving in discrete steps along the cytoskeleton and carrying a molecular load. Single-molecule recordings of motor position along a substrate polymer appear as a stochastic staircase. Recordings of other single molecules, such as F1-ATPase, RNA polymerase, or topoisomerase, have the same appearance. We present a maximum likelihood algorithm that extracts the dwell time sequence from noisy data, and estimates state transition probabilities and the distribution of the motor step size. The algorithm can handle models with uniform or alternating step sizes, and reversible or irreversible kinetics. A periodic Markov model describes the repetitive chemistry of the motor, and a Kalman filter allows one to include models with variable step size and to correct for baseline drift. The data are optimized recursively and globally over single or multiple data sets, making the results objective over the full scale of the data. Local binary algorithms, such as the t-test, do not represent the behavior of the whole data set. Our method is model-based, and allows rapid testing of different models by comparing the likelihood scores. From data obtained with current technology, steps as small as 8 nm can be resolved and analyzed with our method. The kinetic consequences of the extracted dwell sequence can be further analyzed in detail. We show results from analyzing simulated and experimental kinesin and myosin motor data. The algorithm is implemented in the free QuB software.
PubMedId: 16905607
Journal-article: Biophysical journal, vol. 91, 2006
Authors: Lorin S Milescu, Ahmet Yildiz, Paul R Selvin, Frederick Sachs
Abstract: Molecular motors, such as kinesin, myosin, or dynein, convert chemical energy into mechanical energy by hydrolyzing ATP. The mechanical energy is used for moving in discrete steps along the cytoskeleton and carrying a molecular load. High resolution single molecule recordings of motor steps appear as a stochastic sequence of dwells, resembling a staircase. Staircase data can also be obtained from other molecular machines such as F1 -ATPase, RNA polymerase, or topoisomerase. We developed a maximum likelihood algorithm that estimates the rate constants between different conformational states of the protein, including motor steps. We model the motor with a periodic Markov model that reflects the repetitive chemistry of the motor step. We estimated the kinetics from the idealized dwell-sequence by numerical maximization of the likelihood function for discrete-time Markov models. This approach eliminates the need for missed event correction. The algorithm can fit kinetic models of arbitrary complexity, such as uniform or alternating step chemistry, reversible or irreversible kinetics, ATP concentration and mechanical force-dependent rates, etc. The method allows global fitting across stationary and nonstationary experimental conditions, and user-defined a priori constraints on rate constants. The algorithm was tested with simulated data, and implemented in the free QuB software.
PubMedId: 16679362
Journal-article: Biophysical journal, vol. 91, 2006
Authors: Bridget T Jacques-Fricke, Yiqi Seow, Philip A Gottlieb, Frederick Sachs, Timothy M Gomez
Abstract: Ca2+ signals are known to be important regulators of neurite outgrowth and steering. Here we show that inhibiting Ca2+ influx through stretch-activated channels using various compounds, including a highly specific peptide isolated from Grammostola spatulata spider venom (GsMTx4), strongly accelerates the rate of neurite extension on diverse substrata and within the intact spinal cord. Consistent with the presence of stretch-activated channels, we show that Ca2+ influx is triggered by hypotonic solutions, which can be partially blocked by GsMTx4. Finally, chelating local, but not global, Ca2+ signals prevents the acceleration that is normally produced by GsMTx4. Blocking Ca2+ influx through other channel types has little or opposite effects, but release from intracellular stores is required for maximal acceleration. Together, our data suggest that Ca2+ functions at distinct microdomains in growth cones, with influx through mechanosensitive channels acting to inhibit outgrowth in opposition to influx through other plasma membrane channels and release from stores.
PubMedId: 16723522
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 26, 2006
Authors: E. Honore, A. J. Patel, J. Chemin, T. Suchyna, F. Sachs
PubMedId: 16636285
Journal-article: Proceedings of the National Academy of Sciences, vol. 103, pp. 6859-6864, 2006
Authors: Arthur Beyder, Frederick Sachs
Abstract: We developed a mass production fabrication process for making symmetrically supported torsion cantilevers/oscillators with highly compliant springs. These torsion probes offer advantages in atomic force microscopy (AFM) because they are small, have high optical gain, do not warp and can be made with two independent axes. Compared to traditional AFM cantilevers, these probes have higher frequency response, higher Q, lower noise, better optics (since the mirror does not bend) and two data channels. Soft small levers with sub-pN force resolution can resonate cleanly above 10 kHz in water. When fabricated with a ferromagnetic coating on the rigid reflecting pad, they can be driven magnetically or serve as high-resolution magnetometers. Asymmetric levers can be tapping mode probes or high-resolution accelerometers. The dual axis gimbaled probes with two orthogonal axes can operate on a standard AFM with single beam illumination. These probes can be used as self-referencing, drift free, cantilevers where one axis senses the substrate position and the other the sample position. These levers can be optimized for differential contrast or high-resolution friction imaging.
PubMedId: 16750300
Journal-article: Ultramicroscopy, vol. 106, 2005
Authors: Patricia R Chess, Michael A O'Reilly, Fredrick Sachs, Jacob N Finkelstein
Abstract: Mechanical strain is necessary for normal lung growth and development. Individuals with respiratory failure are supported with mechanical ventilation, leading to altered lung growth and injury. Understanding signaling pathways initiated by mechanical strain in lung epithelial cells will help guide development of strategies aimed at optimizing strain-induced lung growth while mitigating ventilator-induced lung injury. To study strain-induced proliferative signaling, focusing on the role of reactive oxidant species (ROS) and p42/44 mitogen-activated protein (MAP) kinase, human pulmonary epithelial H441 and MLE15 cells were exposed to equibiaxial cyclic mechanical strain. ROS were increased within 15 min of strain. N-acetylcysteine inactivated strain-induced ROS and inhibited p42/44 MAP kinase phosphorylation and strain-induced proliferation. PD98059 and UO126, p42/44 MAP kinase inhibitors, blocked strain-induced proliferation. To verify the specificity of p42/44 MAP kinase inhibition, cells were transfected with dominant-negative mitogen-activated protein kinase kinase-1 plasmid DNA. Transfected cells did not proliferate in response to mechanical strain. To determine whether strain-induced tyrosine kinase activity is necessary for strain-induced ROS-p42/44 MAP kinase signaling, genistein, a tyrosine kinase inhibitor, was used. Genistein did not block strain-induced ROS production or p42/44 MAP kinase phosphorylation. Gadolinium, a mechanosensitive calcium channel blocker, blocked strain-induced ROS production and p42/44 MAP kinase phosphorylation but not strain-induced tyrosine phosphorylation. These data support ROS production and p42/44 MAP kinase phosphorylation being involved in a common strain-induced signaling pathway, necessary for strain-induced proliferation in pulmonary epithelial cells, with a parallel strain-induced tyrosine kinase pathway.
PubMedId: 15890751
Journal-article: Journal of applied physiology (Bethesda, Md. : 1985), vol. 99, 2005
Authors: Lorin S Milescu, Gustav Akk, Frederick Sachs
Abstract: We describe a maximum likelihood method for direct estimation of rate constants from macroscopic ion channel data for kinetic models of arbitrary size and topology. The number of channels in the preparation, and the mean and standard deviation of the unitary current can be estimated, and a priori constraints can be imposed on rate constants. The method allows for arbitrary stimulation protocols, including stimuli with finite rise time, trains of ligand or voltage steps, and global fitting across different experimental conditions. The initial state occupancies can be optimized from the fit kinetics. Utilizing arbitrary stimulation protocols and using the mean and the variance of the current reduce or eliminate problems of model identifiability (Kienker, 1989). The algorithm is faster than a recent method that uses the full autocovariance matrix (Celentano and Hawkes, 2004), in part due to the analytical calculation of the likelihood gradients. We tested the method with simulated data and with real macroscopic currents from acetylcholine receptors, elicited in response to brief pulses of carbachol. Given appropriate stimulation protocols, our method chose a reasonable model size and topology.
PubMedId: 15681642
Journal-article: Biophysical journal, vol. 88, 2005
Authors: Li Bao, Frederick Sachs, Gerhard Dahl
Abstract: Connexins form gap junction channels that provide a hydrophilic path between cell interiors. Some connexins, particularly the lens connexins, Cx46 and Cx50 and their orthologs, can form functional hemichannels in nonjunctional membranes. These hemichannels are a nonselective conduit to the extracellular medium and may jeopardize cell survival. The physiological function of hemichannels has remained elusive, but it has been postulated that hemichannels are involved in ATP-release caused by mechanical stimulation. Here we show with single-channel and whole cell electrophysiological studies that Cx46 hemichannels are mechanosensitive, like other families of ion channels and membrane-bound enzymes. The hemichannel response to mechanical stress is bipolar. At negative potentials stress opens the channel, and at positive potentials stress closes it. Physiologically, Cx46 hemichannels may assist accommodation of the ocular lens by providing a transient path for volume flow as the lens changes shape.
PubMedId: 15475518
Journal-article: American journal of physiology. Cell physiology, vol. 287, 2004
Authors: Philip A Gottlieb, Thomas M Suchyna, Lyle W Ostrow, Frederick Sachs
PubMedId: 15379605
Journal Article: Current drug targets. CNS and neurological disorders, vol. 3, 2004
Authors: S Besch, K V Snyder, P C Zhang, F Sachs
PubMedId: 14716076
Journal Article: Cell biochemistry and biophysics, vol. 39, 2004
Authors: Lyle W Ostrow, Frederick Sachs
Abstract: Endothelin (ET) is a potent autocrine mitogen produced by reactive and neoplastic astrocytes. ET has been implicated in the induction of astrocyte proliferation and other transformations engendered by brain pathology, and in promoting the malignant behavior of astrocytomas. Reactive astrocytes containing ET are found in the periphery/penumbra of a wide array of CNS pathologies. Virtually all brain pathology deforms the surrounding parenchyma, either by direct mass effect or edema. Mechanical stress is a well established stimulus for ET production and release by other cell types, but has not been well studied in the brain. However, numerous studies have illustrated that astrocytes can sense mechanical stress and translate it into chemical messages. Furthermore, the ubiquitous reticular meshwork formed by interconnected astrocytes provides an ideal morphology for sensing and responding to mechanical disturbances. We have recently demonstrated stretch-induced ET production by astrocytes in vitro. Inspired by this finding, the purpose of this article is to review the literature on (1) astrocyte mechanosensation, and (2) the endothelin system in astrocytes, and to consider the hypothesis that mechanical induction of the ET system may influence astrocyte functioning in CNS pathophysiology. We conclude by discussing evidence supporting future investigations to determine whether specific inhibition of stretch-activated ion channels may represent a novel strategy for treating or preventing CNS disturbances, as well as the relevance to astrocyte-derived tumors.
PubMedId: 15914254
Journal-article: Brain research. Brain research reviews, vol. 48, 2003
Authors: Kimberly Laskie Ostrow, Aaron Mammoser, Tom Suchyna, Frederick Sachs, Robert Oswald, Shigeru Kubo, Naoyoshi Chino, Philip A Gottlieb
Journal-article: Toxicon, vol. 42, pp. 263-274, 2003
Authors: Weizhen Niu, Frederick Sachs
Journal-article: Progress in Biophysics and Molecular Biology, vol. 82, pp. 121-135, 2003
Authors: Weizhen Niu, Frederick Sachs
PubMedId: 12732273
Journal Article: Progress in biophysics and molecular biology, vol. 82, 2003
Authors: Alexander G. Petrov, Frederick Sachs
Journal-article: Physical Review E, vol. 65, 2002
Authors: Ping-Cheng Zhang, Asbed M. Keleshian, Frederick Sachs
Journal-article: Nature, vol. 413, pp. 428-432, 2001
Authors: Feng Qin, Anthony Auerbach, Frederick Sachs
Journal-article: Biophysical Journal, vol. 79, pp. 1915-1927, 2000
Authors: Thomas M. Suchyna, Janice H. Johnson, Katherine Hamer, Joseph F. Leykam, Douglas A. Gage, Henry F. Clemo, Clive M. Baumgarten, Frederick Sachs
Journal-article: The Journal of General Physiology, vol. 115, pp. 583-598, 2000
Authors: J. Niggel, W. Sigurdson, F. Sachs
Journal-article: Journal of Membrane Biology, vol. 174, pp. 121-134, 2000
Authors: G.C.L. Bett, F. Sachs
Journal-article: Journal of Membrane Biology, vol. 173, pp. 255-263, 2000
Authors: G.C.L. Bett, F. Sachs
Journal-article: The Journal of Membrane Biology, vol. 173, pp. 237, 2000
Authors: Tao Zeng, Glenna C. L. Bett, Frederick Sachs
Journal-article: American Journal of Physiology-Heart and Circulatory Physiology, vol. 278, pp. H548-H557, 2000
Authors: G C Bett, F Sachs
PubMedId: 10667920
Journal Article: The Journal of membrane biology, vol. 173, 2000
Authors: Shinghua Ding, Frederick Sachs
Journal-article: The Journal of Physiology, vol. 522, pp. 199-214, 2000
Authors: S. Ding, F. Sachs
Journal-article: Journal of Membrane Biology, vol. 172, pp. 215-223, 1999
Authors: Frederick Sachs
Journal-article: Biophysical Journal, vol. 77, pp. 682-690, 1999
Authors: Shinghua Ding, Frederick Sachs
Journal-article: The Journal of General Physiology, vol. 113, pp. 695-720, 1999
Authors: Sergei I. Sukharev, Wade J. Sigurdson, Ching Kung, Frederick Sachs
Journal-article: The Journal of General Physiology, vol. 113, pp. 525-540, 1999
Authors: Johannes Akinlaja, Frederick Sachs
Journal-article: Biophysical Journal, vol. 75, pp. 247-254, 1998
Authors: E. J. Luna, A. L. Hitt, D. Shutt, D. Wessels, D. Soll, P. Jay, C. Hug, E. L. Elson, A. Vesley, G. P. Downey, M. Wang, S. M. Block, W. Sigurdson, F. Sachs
Journal-article: The Biological Bulletin, vol. 194, pp. 345-347, 1998
Authors: J. Mosbacher, M. Langer, J.K.H. H??rber, F. Sachs
Journal-article: The Journal of General Physiology, vol. 111, pp. 65-74, 1998
Authors: Frederick Sachs
Journal-article: Nature, vol. 390, pp. 203-203, 1997
Authors: Benyuan Zhang, Wade Sigurdson, Frederick Sachs
Journal-article: Journal of Neuroscience Methods, vol. 76, pp. 151-155, 1997
Authors: Hai Hu, Frederick Sachs
Journal-article: Journal of Molecular and Cellular Cardiology, vol. 29, pp. 1511-1523, 1997
Authors: F. Qin, A. Auerbach, F. Sachs
Journal-article: Proceedings of the Royal Society B: Biological Sciences, vol. 264, pp. 375-383, 1997
Authors: F Sachs
PubMedId: 9210231
Journal Article: Society of General Physiologists series, vol. 52, 1997
Authors: H. Hu, F. Sachs
Journal-article: Journal of Membrane Biology, vol. 154, pp. 205-216, 1996
Authors: M. Zabel, B. S. Koller, F. Sachs, M. R. Franz
Journal-article: Cardiovascular Research, vol. 32, pp. 120-130, 1996
Authors: F. Qin, A. Auerbach, F. Sachs
Journal-article: Biophysical Journal, vol. 70, pp. 264-280, 1996
Authors: Frederick Sachs
Journal-article: Pfl???gers Archiv European Journal of Physiology, vol. 429, pp. 434-435, 1995
Authors: S. L. Diamond, F. Sachs, W. J. Sigurdson
Journal-article: Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 14, pp. 2000-2006, 1994
Authors: F. Sachs, F. Qin
Journal-article: Biophysical Journal, vol. 65, pp. 1101-1107, 1993
Authors: A. Ruknudin, F. Sachs, J. O. Bustamante
Journal-article: American Journal of Physiology-Heart and Circulatory Physiology, vol. 264, pp. H960-H972, 1993
Authors: Xian-Cheng Yang, Frederick Sachs
Book-chapter: Nonselective Cation Channels, pp. 79-92, 1993
Authors: Charles L. Bowman, Jiu-Ping Ding, Frederick Sachs, Masahiro Sokabe
Journal-article: Brain Research, vol. 584, pp. 272-286, 1992
Authors: F Sachs, W Sigurdson, A Ruknudin, C Bowman
PubMedId: 17835497
Journal-article: Science (New York, N.Y.), vol. 253, 1991
Authors: J.P. Ding, R.J. Salvi, F. Sachs
Journal-article: Hearing Research, vol. 56, pp. 19-28, 1991
Authors: Ye Chen Izu, Frederick Sachs
Journal-article: Pfl???gers Archiv European Journal of Physiology, vol. 419, pp. 218-220, 1991
Authors: F. SACHS, W. SIGURDSON, A. RUKNUDIN, C. BOWMAN
Journal-article: Science, pp. 800-801, 1991
Authors: F. Sachs
Journal-article: Molecular and Cellular Biochemistry, vol. 104, 1991
Authors: F. Sachs
Book-chapter: Molecular Mechanisms of Cellular Growth, pp. 57-60, 1991
Authors: F Sachs
PubMedId: 1717821
Journal Article: Molecular and cellular biochemistry, vol. 104, 1991
Authors: M. Sokabe, F. Sachs, Z.Q. Jing
Journal-article: Biophysical Journal, vol. 59, pp. 722-728, 1991
Authors: Zhongqi Jing, Frederick Sachs
Journal-article: Ultramicroscopy, vol. 35, pp. 37-43, 1991
Authors: J O Bustamante, A Ruknudin, F Sachs
PubMedId: 1715454
Journal Article: Journal of cardiovascular pharmacology, vol. 17 Suppl 2, 1991
Authors: X C Yang, F Sachs
Journal-article: The Journal of Physiology, vol. 431, pp. 103-122, 1990
Authors: Frederick Sachs, Masahiro Sokabe
Journal-article: Neuroscience Research Supplements, vol. 12, pp. S1-S4, 1990
Authors: F Sachs, M Sokabe
PubMedId: 1700845
Journal Article: Neuroscience research. Supplement : the official journal of the Japan Neuroscience Society, vol. 12, 1990
Authors: Xiaoqing Gao, Frederick Sachs
Journal-article: Journal of Neuroscience Methods, vol. 28, pp. 225-227, 1989
Authors: X. Yang, F Sachs
Journal-article: Science, vol. 243, pp. 1068-1071, 1989
Authors: ANTHONY AUERBACH, FREDERICK SACHS, JAMES NEIL, RICHARD McGARRIGLE
Book-chapter: Neuroendocrine Peptide Methodology, pp. 503-518, 1989
Authors: FREDERICK SACHS, ANTHONY AUERBACH
Book-chapter: Neuroendocrine Peptide Methodology, pp. 451-479, 1989
Authors: F Sachs
PubMedId: 2460290
Journal Article: Critical reviews in biomedical engineering, vol. 16, 1988
Authors: Frederick Sachs
Journal-article: Membrane Biochemistry, vol. 6, pp. 173-195, 1986
Authors: F Sachs
PubMedId: 2427917
Journal Article: Membrane biochemistry, vol. 6, 1986
Authors: Anthony Auerbach, Frederick Sachs, James Neil, Richard McGarrigle
Book-chapter: Methods in Enzymology, pp. 190-206, 1986
Authors: A Auerbach, F Sachs, J Neil, R McGarrigle
PubMedId: 3713522
Journal Article: Methods in enzymology, vol. 124, 1986
Authors: F Guharay, F Sachs
Journal-article: The Journal of Physiology, vol. 363, pp. 119-134, 1985
Authors: A Auerbach, F Sachs
PubMedId: 6331285
Journal-article: Annual review of biophysics and bioengineering, vol. 13, 1984
Authors: F Guharay, F Sachs
Journal-article: The Journal of Physiology, vol. 352, pp. 685-701, 1984
Authors: A. Auerbach, F. Sachs
Journal-article: Biophysical Journal, vol. 45, pp. 187-198, 1984
Authors: V. Demjannenko, F. Sachs
Journal-article: Medical & Biological Engineering & Computing, vol. 21, pp. 650-653, 1983
Authors: A. Auerbach, F. Sachs
Journal-article: Biophysical Journal, vol. 42, pp. 1-10, 1983
Authors: Frederick Sachs, Anthony Auerbach
Book-chapter: Methods in Enzymology, pp. 147-176, 1983
Authors: F Sachs, A Auerbach
PubMedId: 6321886
Journal Article: Methods in enzymology, vol. 103, 1983
Authors: F. Sachs, J. Neil, N. Barkakati
Journal-article: Pfl???gers Archiv European Journal of Physiology, vol. 395, pp. 331-340, 1982
Authors: V. Demjanenko, F. Sachs
Journal-article: Medical & Biological Engineering & Computing, vol. 20, pp. 65-69, 1982
Authors: F. Sachs, P. Specht
Journal-article: Medical & Biological Engineering & Computing, vol. 19, pp. 316-320, 1981
Authors: F. Sachs, R. McGarrigle
Journal-article: Journal of Neuroscience Methods, vol. 3, pp. 151-157, 1980
Authors: Frederick Sachs
Journal-article: Journal of Neuroscience Methods, vol. 2, pp. 267-271, 1980
Authors: Deborah J. Nelson, Frederick Sachs
Journal-article: Nature, vol. 282, pp. 861-863, 1979
Authors: D J Nelson, F Sachs
PubMedId: 514365
Journal Article: Nature, vol. 282, 1979
Authors: F. Sachs, H. Lecar
Journal-article: Biophysical Journal, vol. 17, pp. 129-143, 1977
Authors: Frederick Sachs
Journal-article: The Journal of Membrane Biology, vol. 28, pp. 373-399, 1976
Authors: Frederick Sachs, Richard D. Feinman
Journal-article: Thrombosis Research, vol. 8, pp. 43-50, 1976
Authors: F. SACHS, H. LECAR
Journal-article: Nature New Biology, vol. 246, pp. 214-216, 1973
Authors: A Bortoff, F Sachs
Journal-article: American Journal of Physiology-Legacy Content, vol. 218, pp. 576-581, 1970
Authors: F. Sachs, C. E. Morris
Book-chapter: Reviews of Physiology, Biochemistry and Pharmacology, pp. 1-77,
Presented by CUBS © 2024