首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 390 毫秒
1.
The mammalian inner ear separates sounds by their frequency content, and this separation underlies important properties of human hearing, including our ability to understand speech in noisy environments. Studies of genetic disorders of hearing have demonstrated a link between frequency selectivity and wave properties of the tectorial membrane (TM). To understand these wave properties better, we developed chemical manipulations that systematically and reversibly alter TM stiffness and viscosity. Using microfabricated shear probes, we show that (i) reducing pH reduces TM stiffness with little change in TM viscosity and (ii) adding PEG increases TM viscosity with little change in TM stiffness. By applying these manipulations in measurements of TM waves, we show that TM wave speed is determined primarily by stiffness at low frequencies and by viscosity at high frequencies. Both TM viscosity and stiffness affect the longitudinal spread of mechanical excitation through the TM over a broad range of frequencies. Increasing TM viscosity or decreasing stiffness reduces longitudinal spread of mechanical excitation, thereby coupling a smaller range of best frequencies and sharpening tuning. In contrast, increasing viscous loss or decreasing stiffness would tend to broaden tuning in resonance-based TM models. Thus, TM wave and resonance mechanisms are fundamentally different in the way they control frequency selectivity.The sharp frequency selectivity of auditory nerve fiber responses to sound is a hallmark of mammalian cochlear function. This remarkable signal processing originates in the mechanical stage of the cochlear signal processing chain (17), as evidenced by measured motions and mechanical properties of the basilar membrane (BM) (29) and tectorial membrane (TM) (1024). Although the hydromechanical mechanisms underlying BM motions have been characterized based on experimental and theoretical studies, the mechanisms underlying TM motions remain unclear.The TM is an acellular matrix that overlies the hair bundles of sensory receptor cells. Based on its strategic position above the organ of Corti, conventional cochlear models (2529) have implicated local mechanical properties (i.e., mass, stiffness) of the TM in stimulating the sensory hair bundles of hair cells and in cochlear tuning. Recent dynamic measurements of the TM, in vitro (17, 3033) and in vivo (34), suggest that the TM supports longitudinal coupling, with large spatial extents across a broad range of frequencies. This longitudinal coupling manifests in the form of propagating traveling waves that are thought to contribute to hearing mechanisms (17, 21, 30, 3540). Genetic modification studies provide further support that the spatial extent of TM waves may play a significant role in cochlear tuning (30, 32). Although these measurements, models, and genetic modification studies have confirmed the importance of TM mechanical properties in hearing, they have not isolated the distinct roles of TM stiffness and viscosity in generating longitudinally propagating traveling waves of the TM.To understand the contributions of TM material properties on traveling waves better, we developed chemical manipulations to alter the stiffness and viscosity of the TM selectively and reversibly. Because the TM is poroelastic (32, 41), we expect that changes in bath composition can have a direct effect on the mechanical properties of the TM mechanical matrix and its interstitial fluid, which makes up 97% of TM wet weight (42). The addition of PEG has previously been shown to generate an osmotic response that could be accounted for by the permeability of these molecules through the matrix rather than by direct changes to the matrix itself (41). In contrast, changing bath pH has little effect on the osmotic pressure or viscosity of the bath but has been shown to have a direct effect on the macromolecular matrix (43). In this paper, we apply these physicochemical manipulations to alter TM material properties reversibly, and thereby probe their role in controlling longitudinal spread of excitation through the TM.  相似文献   

2.
At our body surface, the epidermis absorbs UV radiation. UV overexposure leads to sunburn with tissue injury and pain. To understand how, we focus on TRPV4, a nonselective cation channel highly expressed in epithelial skin cells and known to function in sensory transduction, a property shared with other transient receptor potential channels. We show that following UVB exposure mice with induced Trpv4 deletions, specifically in keratinocytes, are less sensitive to noxious thermal and mechanical stimuli than control animals. Exploring the mechanism, we find that epidermal TRPV4 orchestrates UVB-evoked skin tissue damage and increased expression of the proalgesic/algogenic mediator endothelin-1. In culture, UVB causes a direct, TRPV4-dependent Ca2+ response in keratinocytes. In mice, topical treatment with a TRPV4-selective inhibitor decreases UVB-evoked pain behavior, epidermal tissue damage, and endothelin-1 expression. In humans, sunburn enhances epidermal expression of TRPV4 and endothelin-1, underscoring the potential of keratinocyte-derived TRPV4 as a therapeutic target for UVB-induced sunburn, in particular pain.The surface epithelium (epidermis) of skin provides barrier protection against dehydration and the potentially harmful external environment (1). Accordingly, skin is the site of first interaction between ambient environment and immunologically competent organismal structures, and also the site for sentient responses (2). Sensory neurons in the dorsal root ganglia (DRG) and trigeminal ganglia (TG) are endowed with sensory transduction capacity for heat, cold, mechanical cues, itch, and pain, and their axons directly interface with skin epithelium (24).Against a background of suggestive findings (2, 57), we wondered whether the epidermis as a “forefront” of sensory signaling may function in sensitizing pain transduction in response to naturally occurring irritating cues. To elucidate mechanisms, we used a mouse sunburn model and induced a state of lowered sensory thresholds associated with tissue injury caused by UV radiation (810). UV-sunburn-evoked lowering of sensory thresholds shares major hallmarks of pathological pain, a valuable feature of this model. Skin tissue injury caused by UVB has been elucidated to be mediated by cytokines and chemokines, known from immunological responses, such as IL-1β and IL-6, which are also known to cause and facilitate pain (1119). Another more recent study identified a proinflammatory chemokine, CXCL5, as proalgesic in response to UVB overexposure of rat and human skin (20). An exciting new arena pertaining to molecular mechanisms of the skin’s response to noxious UV was recently opened by an elegant study that reported the role of UVB-mediated damage to noncoding RNA molecules in the skin (21). Unraveling a molecular mechanism, the Toll-like receptor 3 gene was found critical in signaling the proinflammatory actions of the UVB-damaged noncoding RNA molecules. However, this study focused on molecular mechanisms of acute inflammation in the skin.We intended to identify pain mechanisms that mediate the pain associated with UVB-mediated tissue injury. Pain in response to external environmental cues has been understood better because of scientific progress in the field of transient receptor potential (TRP) ion channels that have been found responsive to such cues, and which were found expressed in DRG and TG peripheral sensory neurons, which are the cells believed to be the primary transducers. Indeed, TRPV1, one of the founding members of the TRPV channel subfamily, has been identified as relevant for pain, including pathological pain, response to thermal cues, and most recently for itch (2231). However, TRPA1 (transient receptor potential ion channel, ankyrin subfamily, family member #1) and TRPM8 seem to be involved in transduction of pain-inducing stimuli as well (3236).Also a family member of the TRPV subfamily, TRPV4 is a multimodally activated, nonselective cation channel that is involved in physiological pain evoked by osmotic and mechanical, but not thermal, cues (3740). For pathological pain, it is relevant for inflammation- and nerve-damage-induced pain sensitization (4143). Of note, Trpv4−/− mice exhibit impaired skin-barrier function (44, 45). That said, TRPV4 is expressed in a number of different cell types, including robust expression in epidermal keratinocytes and also is detectable in skin-innervating sensory neurons. This “dual-location expression” of TRPV4 leaves the cellular mechanisms involved in the channel’s function and the functional contribution of environment-exposed keratinocytes vs. skin-innervating sensory neurons unclear.Against this background of dual-location TRPV4 expression and the role of TRPV4 in inflammatory and neuropathic pain, we now address whether epidermally derived TRPV4 is pathophysiologically relevant in sunburn pain and tissue damage. Using Trpv4 gene-targeted mice, selectively inducing targeting in postnatal keratinocytes, and topically applying selective TRPV4 inhibitors, we demonstrate that epidermal TRPV4 plays a prominent, hitherto unrecognized role in UVB-evoked skin tissue damage and pain of sunburn.  相似文献   

3.
The apical domain of embryonic (radial glia) and adult (B1 cells) neural stem cells (NSCs) contains a primary cilium. This organelle has been suggested to function as an antenna for the detection of morphogens or growth factors. In particular, primary cilia are essential for Hedgehog (Hh) signaling, which plays key roles in brain development. Their unique location facing the ventricular lumen suggests that primary cilia in NSCs could play an important role in reception of signals within the cerebrospinal fluid. Surprisingly, ablation of primary cilia using conditional alleles for genes essential for intraflagellar transport [kinesin family member 3A (Kif3a) and intraflagellar transport 88 (Ift88)] and Cre drivers that are activated at early [Nestin; embryonic day 10.5 (E10.5)] and late [human glial fibrillary acidic protein (hGFAP); E13.5] stages of mouse neural development resulted in no apparent developmental defects. Neurogenesis in the ventricular–subventricular zone (V-SVZ) shortly after birth was also largely unaffected, except for a restricted ventral domain previously known to be regulated by Hh signaling. However, Kif3a and Ift88 genetic ablation also disrupts ependymal cilia, resulting in hydrocephalus by postnatal day 4. To directly study the role of B1 cells’ primary cilia without the confounding effects of hydrocephalus, we stereotaxically targeted elimination of Kif3a from a subpopulation of radial glia, which resulted in ablation of primary cilia in a subset of B1 cells. Again, this experiment resulted in decreased neurogenesis only in the ventral V-SVZ. Primary cilia ablation led to disruption of Hh signaling in this subdomain. We conclude that primary cilia are required in a specific Hh-regulated subregion of the postnatal V-SVZ.The primary cilium, a minute elongated organelle with a (9+0) microtubular cytoskeleton (axoneme) on the surface of most cells, is essential for signal transduction and particularly for Hedgehog (Hh) signaling (14). The primary cilium, therefore, has very important functions during vertebrate development (5, 6), including the development of the central nervous system (79). Primary cilia are required for the expansion of progenitor pool during cerebellar development (10, 11) and in the formation of neural stem cells (NSCs) and progenitors in the adult hippocampus (1214). Moreover, it has been shown that primary cilia regulate dendritic refinement and synaptic integration of adult-born hippocampal neurons (15). Recent evidence also shows that Arl13b in primary cilia is essential for the early polarization of the neuroepithelium and the formation of radial glia (16). In addition, primary cilia and Arl13b regulate migration and placement of interneurons in the developing cerebral cortex (17, 18).The walls of the lateral ventricles retain an active germinal niche in the ventricular–subventricular zone (V-SVZ) that continues generating neurons and glial cells in the postnatal brain of many mammals (19). The astroglia-like NSCs (B1 cells) give rise to intermediate progenitor cells (C cells), which in turn generate neuroblasts (A cells) (2022). These young neurons migrate along the rostral migratory stream to the olfactory bulb (OB). B1 cells retain epithelial characteristics, including an apical domain that contacts the lateral ventricle (23). This apical process contains a primary cilium and is surrounded by multiciliated ependymal (E1) cells in a pinwheel-like organization (23). Given their location and the important functions that primary cilia have in the processing of extracellular signals, B1 cells’ primary cilia could have key roles in the reception of ventricular signals for the regulation of adult neurogenesis (24, 25). However, the function of B1 cells’ primary cilia remains unknown. Genetically ablating primary cilia—by removing essential components of the intraflagellar transport (IFT) system (26)—inevitably eliminate the motile cilia of E1 cells, resulting in disruption of cerebrospinal fluid (CSF) flow and hydrocephalus. Because E1 cells and CSF are thought to play important roles in the regulation of B1 cell proliferation, it is not possible to dissociate non–cell-autonomous effects of disruption of ependymal cilia from direct effects of primary cilia removal in B1 cells.Here we used various approaches to genetically ablate primary cilia in NSCs at different developmental stages and in different locations. Surprisingly, we found that primary cilia removal during fetal development had strikingly little effect on the development of the telencephalon. During early postnatal life, primary cilia were also dispensable in most B1 cells, but were essential in a specific Hh-regulated subdomain of the V-SVZ. Our results suggest that primary cilia function is tightly linked to Hh signaling within a restricted domain of the postnatal neurogenic region.  相似文献   

4.
Proalgesic sensitization of peripheral nociceptors in painful syndromes is a complex molecular process poorly understood that involves mobilization of thermosensory receptors to the neuronal surface. However, whether recruitment of vesicular thermoTRP channels is a general mechanism underlying sensitization of all nociceptor types or is subtype-specific remains controversial. We report that sensitization-induced Ca2+-dependent exocytotic insertion of transient receptor potential vanilloid 1 (TRPV1) receptors to the neuronal plasma membrane is a mechanism specifically used by peptidergic nociceptors to potentiate their excitability. Notably, we found that TRPV1 is present in large dense-core vesicles (LDCVs) that were mobilized to the neuronal surface in response to a sensitizing insult. Deletion or silencing of calcitonin-gene–related peptide alpha (αCGRP) gene expression drastically reduced proalgesic TRPV1 potentiation in peptidergic nociceptors by abrogating its Ca2+-dependent exocytotic recruitment. These findings uncover a context-dependent molecular mechanism of TRPV1 algesic sensitization and a previously unrecognized role of αCGRP in LDCV mobilization in peptidergic nociceptors. Furthermore, these results imply that concurrent secretion of neuropeptides and channels in peptidergic C-type nociceptors facilitates a rapid modulation of pain signaling.Transient receptor potential vanilloid 1 (TRPV1) is a nonspecific cationic channel activated by capsaicin, noxious heat, acid pH, voltage, and membrane-derived lipids (1). TRPV1 upregulation in sensory neurons is a key element in pain development and maintenance of several pathological chronic conditions (25). Its contribution to thermal hyperalgesia has been well-defined through pharmacological and knockout studies (6, 7). TRPV1 channel is subject to a complex regulation by proalgesics that potentiate its activity through modulation of its gating properties (8, 9) and/or by increasing the expression of new channels into the neuronal surface (10, 11). Whether these mechanisms are a general means to sensitize TRPV1 in all nociceptor subpopulations or are cell-type–specific remains contentious.TRPV1 is widely expressed in peptidergic and nonpeptidergic nociceptors from the neonatal and adult rat peripheral nervous system (12, 13). In mice, TRPV1 is transiently present in a wider range of sensory neurons during development, but its expression gradually becomes restricted to the peptidergic subpopulation in adult animals (14, 15). The presence of TRPV1 in C-type peptidergic nociceptors is readily evidenced by its colocalization with the main proinflammatory neuropeptides calcitonin-gene–related peptide alpha (αCGRP) and substance P (SP) (16, 17). αCGRP and SP are pivotal to develop and maintain neurogenic pain and inflammation as genetic ablation of these peptides results in pain resistance (1821). These neuropeptides are selectively stored in large dense-core vesicles (LDCVs) and are released in response to proalgesic stimuli through a regulated, Ca2+-dependent, and SNARE-mediated secretory pathway (22). LDCVs can also serve as carriers of a plethora of signaling molecules, including ion channels and receptors that may enable fast modulation of neuronal excitability (23). This finding raises the exciting hypothesis that proalgesic recruitment of TRPV1 channels is a mechanism occurring in peptidergic nociceptors. We used ATP, a well-known proalgesic agent (2426), to investigate the mechanisms underlying TRPV1 sensitization in sensory neurons. We report that ATP-induced TRPV1 potentiation in peptidergic nociceptors specifically involves the exocytotic mobilization of new channels to the cell surface. Notably, knockout of αCGRP expression inhibited the ATP-evoked TRPV1 delivery to the neuronal surface and prevented ATP-induced in vivo thermal hyperalgesia. Thus, algesic-induced exocytosis of TRPV1 channels in peptidergic nociceptors is a central mechanism in pain signaling.  相似文献   

5.
Mechanosensitive ion channels are sensors probing membrane tension in all species; despite their importance and vital role in many cell functions, their gating mechanism remains to be elucidated. Here, we determined the conditions for releasing intact mechanosensitive channel of large conductance (MscL) proteins from their detergents in the gas phase using native ion mobility–mass spectrometry (IM-MS). By using IM-MS, we could detect the native mass of MscL from Escherichia coli, determine various global structural changes during its gating by measuring the rotationally averaged collision cross-sections, and show that it can function in the absence of a lipid bilayer. We could detect global conformational changes during MscL gating as small as 3%. Our findings will allow studying native structure of many other membrane proteins.One of the best candidates to explore the gating of mechanosensitive channels is the mechanosensitive channel of large conductance (MscL) from Escherichia coli. The crystal structure of MscL in its closed/nearly closed state from Mycobacterium tuberculosis revealed this channel as a homopentamer (1). Each subunit has a cytoplasmic N- and C-terminal domain as well as two α-helical transmembrane (TM) domains, TM1 and TM2, which are connected by a periplasmic loop. The five TM1 helices form the pore and the more peripheral TM2 helices interact with the lipid bilayer.MscL detects changes in membrane tension invoked by a hypoosmotic shock and couples the tension sensing directly to large conformational changes (1, 2). On the basis of a large body of structural and theoretical data, numerous gating models of MscL have been proposed (39). These models agree upon (i) the hydrophobic pore constriction of the channel and (ii) the channel opens by an iris-like rotation—i.e., a tilting and outward movement of transmembrane helices that make the channel wider and shorter (5). This mechanism is supported by patch-clamp (10), disulfide cross-linking (11), FRET spectroscopy (12), and site-directed spin labeling EPR experiments (6, 7), as well as computational studies (1315). So far, direct experimental results have only been observed for short-range local structural changes, and no measure of the overall global structural changes during channel gating have been reported. Because there is no crystal structure available for the open MscL channel, elucidating overall global structural changes from the onset of channel activation is of utmost importance for our understanding of the gating mechanism of mechanosensitive channels. Here, we provide direct experimental evidence for the key areal changes occurring during channel gating by combining our ability to activate MscL in a controlled manner to different subopen states (16) with a native ion mobility-mass spectrometry (IM-MS) approach.  相似文献   

6.
The capsaicin receptor transient receptor potential cation channel vanilloid 1 (TRPV1) is activated by various noxious stimuli, and the stimuli are converted into electrical signals in primary sensory neurons. It is believed that cation influx through TRPV1 causes depolarization, leading to the activation of voltage-gated sodium channels, followed by the generation of action potential. Here we report that the capsaicin-evoked action potential could be induced by two components: a cation influx-mediated depolarization caused by TRPV1 activation and a subsequent anion efflux-mediated depolarization via activation of anoctamin 1 (ANO1), a calcium-activated chloride channel, resulting from the entry of calcium through TRPV1. The interaction between TRPV1 and ANO1 is based on their physical binding. Capsaicin activated the chloride currents in an extracellular calcium-dependent manner in HEK293T cells expressing TRPV1 and ANO1. Similarly, in mouse dorsal root ganglion neurons, capsaicin-activated inward currents were inhibited significantly by a specific ANO1 antagonist, T16Ainh-A01 (A01), in the presence of a high concentration of EGTA but not in the presence of BAPTA [1,2-bis(o-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid]. The generation of a capsaicin-evoked action potential also was inhibited by A01. Furthermore, pain-related behaviors in mice treated with capsaicin, but not with αβ-methylene ATP, were reduced significantly by the concomitant administration of A01. These results indicate that TRPV1–ANO1 interaction is a significant pain-enhancing mechanism in the peripheral nervous system.When calcium ions enter cells through ion channels or transporters, they can initiate a variety of reactions, either as free calcium ions or after their binding by specific calcium-binding proteins (1). One such important reaction is the activation of calcium-binding proteins by calcium nanodomains of the calcium pathways (2). In this regard, some transient receptor potential (TRP) channels have high calcium permeability (3), and it is likely that they activate calcium-binding proteins in the cytosol or plasma membrane. Indeed, TRP vanilloid 4 (TRPV4), a thermosensitive TRP channel (reportedly an osmo- or mechano-sensor) (48) and anoctamin 1 (ANO1; a calcium-activated chloride channel) (911) function as a complex in epithelial cells of the choroid plexus (12). Upon entering choroid plexus epithelial cells, calcium activates ANO1, leading to chloride efflux. Although the interaction between TRP channels and anoctamins could work in a variety of ways (12), the direction of chloride movement is determined simply by the relationship between chloride equilibrium potentials and membrane potentials, depending on the intracellular chloride concentrations (13). This concept prompted us to pursue other interactions between TRP channels and anoctamins. We focused on primary sensory neurons because activation of chloride channels in sensory neurons causes chloride efflux and depolarization, as the result of the high intracellular chloride concentrations (14, 15).TRPV1 senses various noxious stimuli in primary sensory neurons, leading to pain perception through the generation of action potentials upon the activation of voltage-gated sodium channels. Mammalian TRPV1 is activated by noxious heat, acid, and many chemical compounds including capsaicin (1618). The calcium permeability of TRPV1 is more than 10 times that of sodium, suggesting that TRPV1 could activate anoctamins readily, leading to further depolarization. ANO1 plays an important role in nociception in primary sensory neurons (19), and bradykinin-induced and neuropathic pain-related behaviors were reduced in ANO1 conditional-knockout mice (20, 21), suggesting that interaction between the two proteins could strongly enhance nociceptive signals.  相似文献   

7.
Rickettsiae are responsible for some of the most devastating human infections. A high infectivity and severe illness after inhalation make some rickettsiae bioterrorism threats. We report that deletion of the exchange protein directly activated by cAMP (Epac) gene, Epac1, in mice protects them from an ordinarily lethal dose of rickettsiae. Inhibition of Epac1 suppresses bacterial adhesion and invasion. Most importantly, pharmacological inhibition of Epac1 in vivo using an Epac-specific small-molecule inhibitor, ESI-09, completely recapitulates the Epac1 knockout phenotype. ESI-09 treatment dramatically decreases the morbidity and mortality associated with fatal spotted fever rickettsiosis. Our results demonstrate that Epac1-mediated signaling represents a mechanism for host–pathogen interactions and that Epac1 is a potential target for the prevention and treatment of fatal rickettsioses.Rickettsiae are responsible for some of the most devastating human infections (14). It has been forecasted that temperature increases attributable to global climate change will lead to more widespread distribution of rickettsioses (5). These tick-borne diseases are caused by obligately intracellular bacteria of the genus Rickettsia, including Rickettsia rickettsii, the causative agent of Rocky Mountain spotted fever (RMSF) in the United States and Latin America (2, 3), and Rickettsia conorii, the causative agent of Mediterranean spotted fever endemic to southern Europe, North Africa, and India (6). A high infectivity and severe illness after inhalation make some rickettsiae (including Rickettsia prowazekii, R. rickettsii, Rickettsia typhi, and R. conorii) bioterrorism threats (7). Although the majority of rickettsial infections can be controlled by appropriate broad-spectrum antibiotic therapy if diagnosed early, up to 20% of misdiagnosed or untreated (1, 3) and 5% of treated RMSF cases (8) result in a fatal outcome caused by acute disseminated vascular endothelial infection and damage (9). Fatality rates as high as 32% have been reported in hospitalized patients diagnosed with Mediterranean spotted fever (10). In addition, strains of R. prowazekii resistant to tetracycline and chloramphenicol have been developed in laboratories (11). Disseminated endothelial infection and endothelial barrier disruption with increased microvascular permeability are the central features of SFG rickettsioses (1, 2, 9). The molecular mechanisms involved in rickettsial infection remain incompletely elucidated (9, 12). A comprehensive understanding of rickettsial pathogenesis and the development of novel mechanism-based treatment are urgently needed.Living organisms use intricate signaling networks for sensing and responding to changes in the external environment. cAMP, a ubiquitous second messenger, is an important molecular switch that translates environmental signals into regulatory effects in cells (13). As such, a number of microbial pathogens have evolved a set of diverse virulence-enhancing strategies that exploit the cAMP-signaling pathways of their hosts (14). The intracellular functions of cAMP are predominantly mediated by the classic cAMP receptor, protein kinase A (PKA), and the more recently discovered exchange protein directly activated by cAMP (Epac) (15). Thus, far, two isoforms, Epac1 and Epac2, have been identified in humans (16, 17). Epac proteins function by responding to increased intracellular cAMP levels and activating the Ras superfamily small GTPases Ras-proximate 1 and 2 (Rap1 and Rap2). Accumulating evidence demonstrates that the cAMP/Epac1 signaling axis plays key regulatory roles in controlling various cellular functions in endothelial cells in vitro, including cell adhesion (1821), exocytosis (22), tissue plasminogen activator expression (23), suppressor of cytokine signaling 3 (SOCS-3) induction (2427), microtubule dynamics (28, 29), cell–cell junctions, and permeability and barrier functions (3037). Considering the critical importance of endothelial cells in rickettsioses, we examined the functional roles of Epac1 in rickettsial pathogenesis in vivo, taking advantage of the recently generated Epac1 knockout mouse (38) and Epac-specific inhibitors (39, 40) generated from our laboratory. Our studies demonstrate that Epac1 plays a key role in rickettsial infection and represents a therapeutic target for fatal rickettsioses.  相似文献   

8.
Mechanical loading of joints plays a critical role in maintaining the health and function of articular cartilage. The mechanism(s) of chondrocyte mechanotransduction are not fully understood, but could provide important insights into new physical or pharmacologic therapies for joint diseases. Transient receptor potential vanilloid 4 (TRPV4), a Ca2+-permeable osmomechano-TRP channel, is highly expressed in articular chondrocytes, and loss of TRPV4 function is associated with joint arthropathy and osteoarthritis. The goal of this study was to examine the hypothesis that TRPV4 transduces dynamic compressive loading in articular chondrocytes. We first confirmed the presence of physically induced, TRPV4-dependent intracellular Ca2+ signaling in agarose-embedded chondrocytes, and then used this model system to study the role of TRPV4 in regulating the response of chondrocytes to dynamic compression. Inhibition of TRPV4 during dynamic loading prevented acute, mechanically mediated regulation of proanabolic and anticatabolic genes, and furthermore, blocked the loading-induced enhancement of matrix accumulation and mechanical properties. Furthermore, chemical activation of TRPV4 by the agonist GSK1016790A in the absence of mechanical loading similarly enhanced anabolic and suppressed catabolic gene expression, and potently increased matrix biosynthesis and construct mechanical properties. These findings support the hypothesis that TRPV4-mediated Ca2+ signaling plays a central role in the transduction of mechanical signals to support cartilage extracellular matrix maintenance and joint health. Moreover, these insights raise the possibility of therapeutically targeting TRPV4-mediated mechanotransduction for the treatment of diseases such as osteoarthritis, as well as to enhance matrix formation and functional properties of tissue-engineered cartilage as an alternative to bioreactor-based mechanical stimulation.Articular cartilage is the dense connective tissue that lines the surfaces of diarthrodial joints and provides a low-friction surface for joint loading and articulation. The extracellular matrix (ECM) of articular cartilage is primarily comprised of proteoglycans and type II collagen, in addition to a sparse population of chondrocytes responsible for synthesizing and maintaining this tissue. The mechanical environment of articular cartilage plays an important role in regulating the development and maintenance of the tissue. For example, dynamic compressive loading of cartilage supports ECM biosynthesis (1), whereas abnormal loading, such as disuse, static loading, or altered joint biomechanics, can disrupt ECM homeostasis (2, 3) and lead to osteoarthritis (OA) (4), a degenerative joint disease characterized by an imbalance of chondrocyte anabolic and catabolic activities. Most of the hypotheses on the etiology of OA involve biomechanical loading as a factor (4, 5). As such, understanding chondrocyte mechanotransduction, i.e., how chondrocytes sense and respond to their physical environment, is vital to understanding how OA develops and progresses, and may lead to new treatments for this disease.Chondrocyte mechanotransduction appears to involve the integration and transduction of multiple biophysical signals that arise from joint loading, including direct matrix, cellular, and nuclear strain, hydrostatic pressurization, fluid shear, and changes in tissue osmolarity (6). Ion channels, integrin signaling, and the primary cilia have all been implicated in transducing the external biophysical environment of chondrocytes into electrical and/or chemical intracellular signaling (79). Specifically, intracellular Ca2+ signaling has emerged as a common regulatory mechanism for controlling gene and protein expression (1012).The transient receptor potential vanilloid 4 (TRPV4) channel is a multimodally activated, Ca2+-preferred membrane ion channel widely implicated in transducing external environmental cues into specific metabolic responses via the generation of intracellular Ca2+ ([Ca2+]i) transients (1315). Human TRPV4 mutations that alter channel function are known to disrupt normal skeletal development and joint health (14, 1618), and similarly, targeted deletion of TRPV4 in mice leads to loss of chondrocyte osmotransduction and subsequently, severe joint degeneration (19). TRPV4-mediated Ca2+ signaling has also been shown to enhance chondrogenic gene expression in chondroprogenitor cell lines (20), as well as increase matrix synthesis in chondrocyte-based self-assembled constructs (21). However, the precise role of TRPV4 in transducing and regulating chondrocyte metabolic activity in response to mechanical loading is unclear.The goal of this study was to examine the hypothesis that TRPV4 transduces dynamic compressive loading into signals that regulate cartilage homeostasis. We first confirmed the presence of TRPV4 channels in chondrocyte-laden agarose constructs that produced [Ca2+]i transients in response to hypoosmotic swelling and TRPV4 agonist GSK1016790A (GSK101). The TRPV4 antagonist GSK205 was used to examine the role of this channel in regulating the response of chondrocytes to mechanical loading, whereas the GSK101 and osmotic loading were used to evaluate the effects of TRPV4 activation in the absence of mechanical loading.  相似文献   

9.
10.
11.
12.
13.
14.
Across animal taxa, seminal proteins are important regulators of female reproductive physiology and behavior. However, little is understood about the physiological or molecular mechanisms by which seminal proteins effect these changes. To investigate this topic, we studied the increase in Drosophila melanogaster ovulation behavior induced by mating. Ovulation requires octopamine (OA) signaling from the central nervous system to coordinate an egg’s release from the ovary and its passage into the oviduct. The seminal protein ovulin increases ovulation rates after mating. We tested whether ovulin acts through OA to increase ovulation behavior. Increasing OA neuronal excitability compensated for a lack of ovulin received during mating. Moreover, we identified a mating-dependent relaxation of oviduct musculature, for which ovulin is a necessary and sufficient male contribution. We report further that oviduct muscle relaxation can be induced by activating OA neurons, requires normal metabolic production of OA, and reflects ovulin’s increasing of OA neuronal signaling. Finally, we showed that as a result of ovulin exposure, there is subsequent growth of OA synaptic sites at the oviduct, demonstrating that seminal proteins can contribute to synaptic plasticity. Together, these results demonstrate that ovulin increases ovulation through OA neuronal signaling and, by extension, that seminal proteins can alter reproductive physiology by modulating known female pathways regulating reproduction.Throughout internally fertilizing animals, seminal proteins play important roles in regulating female fertility by altering female physiology and, in some cases, behavior after mating (reviewed in refs. 13). Despite this, little is understood about the physiological mechanisms by which seminal proteins induce postmating changes and how their actions are linked with known networks regulating female reproductive physiology.In Drosophila melanogaster, the suite of seminal proteins has been identified, as have many seminal protein-dependent postmating responses, including changes in egg production and laying, remating behavior, locomotion, feeding, and in ovulation rate (reviewed in refs. 2 and 3). For example, the Drosophila seminal protein ovulin elevates ovulation rate to maximal levels during the 24 h following mating (4, 5), and the seminal protein sex peptide (SP) suppresses female mating receptivity and increases egg-laying behavior for several days after mating (610). However, although a receptor for SP has been identified (11), along with elements of the neural circuit in which it is required (1214), SP’s mechanism of action has not yet been linked to regulatory networks known to control postmating behaviors. Thus, a crucial question remains: how do male-derived seminal proteins interact with regulatory networks in females to trigger postmating responses?We addressed this question by examining the stimulation of Drosophila ovulation by the seminal protein ovulin. In insects, ovulation, defined here as the release of an egg from the ovary to the uterus, is among the best understood reproductive processes in terms of its physiology and neurogenetics (1527). In D. melanogaster, ovulation requires input from neurons in the abdominal ganglia that release the catecholaminergic neuromodulators octopamine (OA) and tyramine (17, 18, 28). Drosophila ovulation also requires an OA receptor, OA receptor in mushroom bodies (OAMB) (19, 20). Moreover, it has been proposed that OA may integrate extrinsic factors to regulate ovulation rates (17). Noradrenaline, the vertebrate structural and functional equivalent to OA (29, 30), is important for mammalian ovulation, and its dysregulation has been associated with ovulation disorders (3138). In this paper we investigate the role of neurons that release OA and tyramine in ovulin’s action. For simplicity, we refer to these neurons as “OA neurons” to reflect the well-established role of OA in ovulation behavior (1620, 22).We investigated how action of the seminal protein ovulin relates to the conserved canonical neuromodulatory pathway that regulates ovulation physiology (3941). We found that ovulin increases ovulation and egg laying through OA neuronal signaling. We also found that ovulin relaxes oviduct muscle tonus, a postmating process that is also mediated by OA neuronal signaling. Finally, subsequent to these effects we detected an ovulin-dependent increase in synaptic sites between OA motor neurons and oviduct muscle, suggesting that ovulin’s stimulation of OA neurons could have increased their synaptic activity. These results suggest that ovulin affects ovulation by manipulating the gain of a neuromodulatory pathway regulating ovulation physiology.  相似文献   

15.
16.
Protein toxins from tarantula venom alter the activity of diverse ion channel proteins, including voltage, stretch, and ligand-activated cation channels. Although tarantula toxins have been shown to partition into membranes, and the membrane is thought to play an important role in their activity, the structural interactions between these toxins and lipid membranes are poorly understood. Here, we use solid-state NMR and neutron diffraction to investigate the interactions between a voltage sensor toxin (VSTx1) and lipid membranes, with the goal of localizing the toxin in the membrane and determining its influence on membrane structure. Our results demonstrate that VSTx1 localizes to the headgroup region of lipid membranes and produces a thinning of the bilayer. The toxin orients such that many basic residues are in the aqueous phase, all three Trp residues adopt interfacial positions, and several hydrophobic residues are within the membrane interior. One remarkable feature of this preferred orientation is that the surface of the toxin that mediates binding to voltage sensors is ideally positioned within the lipid bilayer to favor complex formation between the toxin and the voltage sensor.Protein toxins from venomous organisms have been invaluable tools for studying the ion channel proteins they target. For example, in the case of voltage-activated potassium (Kv) channels, pore-blocking scorpion toxins were used to identify the pore-forming region of the channel (1, 2), and gating modifier tarantula toxins that bind to S1–S4 voltage-sensing domains have helped to identify structural motifs that move at the protein–lipid interface (35). In many instances, these toxin–channel interactions are highly specific, allowing them to be used in target validation and drug development (68).Tarantula toxins are a particularly interesting class of protein toxins that have been found to target all three families of voltage-activated cation channels (3, 912), stretch-activated cation channels (1315), as well as ligand-gated ion channels as diverse as acid-sensing ion channels (ASIC) (1621) and transient receptor potential (TRP) channels (22, 23). The tarantula toxins targeting these ion channels belong to the inhibitor cystine knot (ICK) family of venom toxins that are stabilized by three disulfide bonds at the core of the molecule (16, 17, 2431). Although conventional tarantula toxins vary in length from 30 to 40 aa and contain one ICK motif, the recently discovered double-knot toxin (DkTx) that specifically targets TRPV1 channels contains two separable lobes, each containing its own ICK motif (22, 23).One unifying feature of all tarantula toxins studied thus far is that they act on ion channels by modifying the gating properties of the channel. The best studied of these are the tarantula toxins targeting voltage-activated cation channels, where the toxins bind to the S3b–S4 voltage sensor paddle motif (5, 3236), a helix-turn-helix motif within S1–S4 voltage-sensing domains that moves in response to changes in membrane voltage (3741). Toxins binding to S3b–S4 motifs can influence voltage sensor activation, opening and closing of the pore, or the process of inactivation (4, 5, 36, 4246). The tarantula toxin PcTx1 can promote opening of ASIC channels at neutral pH (16, 18), and DkTx opens TRPV1 in the absence of other stimuli (22, 23), suggesting that these toxin stabilize open states of their target channels.For many of these tarantula toxins, the lipid membrane plays a key role in the mechanism of inhibition. Strong membrane partitioning has been demonstrated for a range of toxins targeting S1–S4 domains in voltage-activated channels (27, 44, 4750), and for GsMTx4 (14, 50), a tarantula toxin that inhibits opening of stretch-activated cation channels in astrocytes, as well as the cloned stretch-activated Piezo1 channel (13, 15). In experiments on stretch-activated channels, both the d- and l-enantiomers of GsMTx4 are active (14, 50), implying that the toxin may not bind directly to the channel. In addition, both forms of the toxin alter the conductance and lifetimes of gramicidin channels (14), suggesting that the toxin inhibits stretch-activated channels by perturbing the interface between the membrane and the channel. In the case of Kv channels, the S1–S4 domains are embedded in the lipid bilayer and interact intimately with lipids (48, 51, 52) and modification in the lipid composition can dramatically alter gating of the channel (48, 5356). In one study on the gating of the Kv2.1/Kv1.2 paddle chimera (53), the tarantula toxin VSTx1 was proposed to inhibit Kv channels by modifying the forces acting between the channel and the membrane. Although these studies implicate a key role for the membrane in the activity of Kv and stretch-activated channels, and for the action of tarantula toxins, the influence of the toxin on membrane structure and dynamics have not been directly examined. The goal of the present study was to localize a tarantula toxin in membranes using structural approaches and to investigate the influence of the toxin on the structure of the lipid bilayer.  相似文献   

17.
18.
19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号