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 62 毫秒
1.
Zinc (Zn2+) is an essential metal in biology, and its bioavailability is highly regulated. Many cell types exhibit fluctuations in Zn2+ that appear to play an important role in cellular function. However, the detailed molecular mechanisms by which Zn2+ dynamics influence cell physiology remain enigmatic. Here, we use a combination of fluorescent biosensors and cell perturbations to define how changes in intracellular Zn2+ impact kinase signaling pathways. By simultaneously monitoring Zn2+ dynamics and kinase activity in individual cells, we quantify changes in labile Zn2+ and directly correlate changes in Zn2+ with ERK and Akt activity. Under our experimental conditions, Zn2+ fluctuations are not toxic and do not activate stress-dependent kinase signaling. We demonstrate that while Zn2+ can nonspecifically inhibit phosphatases leading to sustained kinase activation, ERK and Akt are predominantly activated via upstream signaling and through a common node via Ras. We provide a framework for quantification of Zn2+ fluctuations and correlate these fluctuations with signaling events in single cells to shed light on the role that Zn2+ dynamics play in healthy cell signaling.

Zinc (Zn2+) is an essential metal in biology, with approximately 10% of the proteins encoded by the human genome predicted to bind Zn2+ (1). All cells maintain and regulate a small pool of labile Zn2+ that can be exchanged among Zn2+-binding proteins and Zn2+ biosensors. The concentration of labile Zn2+ in the cytosol, measured in the hundreds of picomolar range (25), falls within the affinity range of many Zn2+ binding proteins, suggesting that under normal conditions many of these proteins will bind Zn2+ and function properly. However, some Zn2+ binders may need higher Zn2+ concentrations in order to function (6). Furthermore, there is growing evidence that mammalian cells experience fluctuations in available Zn2+, and these dynamics have been shown to be important for cell physiology (711).In addition to serving as an important biological cofactor (12), there are increasing examples that Zn2+ also plays a role in biological signaling. Crosstalk has been observed between Zn2+ dynamics and calcium signaling where increases in cytosolic Zn2+ lead to decrease in endoplasmic reticulum (ER) calcium, and conversely, increases in cytosolic calcium change Zn2+ homeostasis in the ER3. Zn2+ sequestration has been shown to block cell cycle progression in both meiotic oocytes (13) and mitotic cells (1416). At a molecular level, picomolar concentrations of Zn2+ potentiate the response of the ryanodine receptor in cardiomyocytes (17). Zn2+ has also been implicated in metabotropic signaling via the G protein–coupled receptor 39 (GPR39 (18)), direct modulation of protein kinase C activity (19), and activation of MAPK kinase signaling pathways in neurons (20), cardiomyocytes (21), and mast cells (7). While the above studies demonstrate that Zn2+ fluctuations influence cellular processes, in many cases the molecular details of how Zn2+ interacts with canonical signaling pathways, second messengers, or serves as a signal itself are unclear. This is especially true for the MAPK pathway.MAPK signaling plays a role in cell proliferation, differentiation, and development and is one of the most well-studied signaling pathways (22). A connection between MAPK signaling and Zn2+ was first reported in 1996 when it was observed that addition of 300 μM ZnCl2 to 3T3 fibroblasts led to increased phosphorylation of ERK1/2 kinases in the MAPK pathway (23). Early studies used epithelial cell lines to study the connection between Zn2+ and ERK signaling (23, 24). More recently, Zn2+ elevation has been demonstrated to increase ERK phosphorylation in dissociated neurons and transformed HT22 cells, where ERK signaling has been linked to synaptic plasticity and memory consolidation (20, 25, 26). The mechanism of ERK activation by Zn2+ remains enigmatic. The leading hypothesis has been that Zn2+ inhibits protein phosphatases, leading to sustained ERK activation. This idea is supported by the observation that ERK-directed phosphatase PP2A activity is reduced when Zn2+ is added to cell lysates (20, 25). Furthermore, it has been demonstrated that certain phosphatases are inhibited by nano- and picomolar concentrations of Zn2+ in vitro, although these phosphatases are not known to directly interact with ERK1/2 (27, 28). However, it is unclear how these bulk in vitro analyses relate to the role of Zn2+ fluctuations in living cells.The connection between Zn2+ and modulation of the MAPK pathway is even more perplexing when examining how Zn2+ influences Ras activity, which acts upstream of Raf-MEK-ERK. Two studies that involved acute perturbation of Zn2+ by adding high concentrations of Zn2+ concluded that Zn2+ promotes Ras activation (29, 30). On the other hand, two genetic screens in Caenorhabditis elegans suggested that Zn2+ inhibits Ras activity (31, 32). While these studies involved different model systems (cell lines versus C. elegans) and different means of altering Zn2+ (acute elevation versus chronic manipulation), it is important to note that there is a lack of consensus on how Zn2+ influences the Ras-Raf-MEK-ERK pathway.In this work we set out to dissect the connection between Zn2+ and ERK in an effort to elucidate the mechanism of activation. Using a combination of kinase translocation reporters and a Förster resonance energy transfer (FRET)-sensor for Zn2+, we quantified the changes in intracellular Zn2+ in response to subtle extracellular perturbations and correlated them directly with changes in kinase activity at the single cell level. We found that while elevated Zn2+ broadly inhibits phosphatase activity to some extent in vitro, in live cells, Zn2+ primarily activates ERK via upstream signaling, suggesting that ERK phosphatase inhibition can’t fully account for the Zn2+-induced increase in ERK activity. Finally, we demonstrate that our Zn2+ conditions activate Ras and Akt signaling along with ERK but that few other kinases are activated, including stress-response kinases JNK, p38, and p53. We therefore propose a mechanism of action where Zn2+ activates ERK and Akt pathways upstream of Ras, while the specific Zn2+-protein interaction remains elusive.  相似文献   

2.
3.
4.
Transmembrane 16A (TMEM16A, anoctamin1), 1 of 10 TMEM16 family proteins, is a Cl channel activated by intracellular Ca2+ and membrane voltage. This channel is also regulated by the membrane phospholipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. We find that two splice variants of TMEM16A show different sensitivity to endogenous PI(4,5)P2 degradation, where TMEM16A(ac) displays higher channel activity and more current inhibition by PI(4,5)P2 depletion than TMEM16A(a). These two channel isoforms differ in the alternative splicing of the c-segment (exon 13). The current amplitude and PI(4,5)P2 sensitivity of both TMEM16A(ac) and (a) are significantly strengthened by decreased free cytosolic ATP and by conditions that decrease phosphorylation by Ca2+/calmodulin-dependent protein kinase II (CaMKII). Noise analysis suggests that the augmentation of currents is due to a rise of single-channel current (i), but not of channel number (N) or open probability (PO). Mutagenesis points to arginine 486 in the first intracellular loop as a putative binding site for PI(4,5)P2, and to serine 673 in the third intracellular loop as a site for regulatory channel phosphorylation that modulates the action of PI(4,5)P2. In silico simulation suggests how phosphorylation of S673 allosterically and differently changes the structure of the distant PI(4,5)P2-binding site between channel splice variants with and without the c-segment exon. In sum, our study reveals the following: differential regulation of alternatively spliced TMEM16A(ac) and (a) by plasma membrane PI(4,5)P2, modification of these effects by channel phosphorylation, identification of the molecular sites, and mechanistic explanation by in silico simulation.

TMEM16A (anoctamin1) plays a wide range of physiological roles in diverse cell types, including contraction of smooth muscle and gastrointestinal motility, secretion of Cl in epithelial cells, detection of noxious heat in nociceptive neurons, modulation of neuronal excitability, and regulation of cell volume (1). TMEM16A channels, from a family of 10 anoctamin proteins (TMEM16A–K), continuously monitor the concentration of intracellular Ca2+ and function as Ca2+-activated Cl channels (24). Several splice variants of TMEM16A generated by combinatorial inclusion or exclusion of four exon segments, a, b, c, and d (57), display unique electrophysiological properties in tissues. Segments a and b lie in the N terminus, and segments c and d lie in the first intracellular loop of TMEM16A. Among the four segments, it is known that b and c help regulate the cytosolic Ca2+ sensitivity and voltage dependence of channel gating. For example, inclusion of the b-segment results in decreased channel sensitivity to intracellular Ca2+ rise, whereas skipping of the c-segment reduces channel activity and also impairs Ca2+ sensitivity (5, 8, 9). In addition to inclusion or skipping of each segment, calmodulin (1013), phosphorylation (1416), protons (1719), and lipids (2027) also impact on the gating of TMEM16A channels.Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] is a key signaling phospholipid in the inner leaflet of the plasma membrane. It acts as a cofactor that regulates many types of ion channels and receptors (2830), and thus depletion of membrane PI(4,5)P2 by the activation of either phospholipase C (PLC) or phosphoinositide 5-phosphatases leads to decreases or increases in gating activity of ion channels. Of the TMEM16 family, TMEM16A, TMEM16B, and TMEM16F are ion channels best known to be modulated by PI(4,5)P2 (2127, 31). Several studies showed that PI(4,5)P2 is required for sustained TMEM16A channel activity and stabilizes the Ca2+-bound open state of the channels (23, 24, 32). Further work located a PI(4,5)P2 regulatory region and demonstrated how PI(4,5)P2 interacts with TMEM16A to regulate channel gating by performing computational simulation. Le et al. (25) proposed that channel activation and desensitization are mediated by two distinct structural modules; one is a PI(4,5)P2-binding module formed by putative PI(4,5)P2-binding residues of TMs 3–5 located near the cytoplasmic membrane interface and another is a Ca2+-binding module of TMs 6–8 involved in the primary opening of the channel pore by Ca2+. Yu et al. (26) identified three key binding sites involved in TMEM16A–PI(4,5)P2 interaction. When PI(4,5)P2 interacts with these binding residues, which form networks with each other, it affects TMEM16A channel gating as a result of the conformational change of TM6.In our study, using exogenous lipid phosphatase tools and mutagenesis, we found that PI(4,5)P2 differentially regulates channel activity depending on the TMEM16A splice variant. In addition, we found that the presence or absence of intracellular ATP is a key determinant of the PI(4,5)P2 sensitivity of TMEM16A. Through structural analysis partly based on a recent cryogenic electron microscopy (cryo-EM) structure of TMEM16A, we also confirmed that phosphorylation of serine 673 by CaMKII allosterically regulates the structure of a PI(4,5)P2 interaction site in the RDR domain of TMEM16A(ac) near to transmembrane segment 3 (TM3). Together, our data reveal a molecular mechanism of TMEM16A channel regulation by PI(4,5)P2, demonstrating that PI(4,5)P2-dependent TMEM16A channel activation can be allosterically modulated by phosphorylation and alternative splicing.  相似文献   

5.
Goblet cells (GCs) are specialized cells of the intestinal epithelium contributing critically to mucosal homeostasis. One of the functions of GCs is to produce and secrete MUC2, the mucin that forms the scaffold of the intestinal mucus layer coating the epithelium and separates the luminal pathogens and commensal microbiota from the host tissues. Although a variety of ion channels and transporters are thought to impact on MUC2 secretion, the specific cellular mechanisms that regulate GC function remain incompletely understood. Previously, we demonstrated that leucine-rich repeat-containing protein 26 (LRRC26), a known regulatory subunit of the Ca2+-and voltage-activated K+ channel (BK channel), localizes specifically to secretory cells within the intestinal tract. Here, utilizing a mouse model in which MUC2 is fluorescently tagged, thereby allowing visualization of single GCs in intact colonic crypts, we show that murine colonic GCs have functional LRRC26-associated BK channels. In the absence of LRRC26, BK channels are present in GCs, but are not activated at physiological conditions. In contrast, all tested MUC2 cells completely lacked BK channels. Moreover, LRRC26-associated BK channels underlie the BK channel contribution to the resting transepithelial current across mouse distal colonic mucosa. Genetic ablation of either LRRC26 or BK pore-forming α-subunit in mice results in a dramatically enhanced susceptibility to colitis induced by dextran sodium sulfate. These results demonstrate that normal potassium flux through LRRC26-associated BK channels in GCs has protective effects against colitis in mice.

The colonic epithelium is composed of a single layer of heterogeneous cells, covered by mucus, that separate the luminal contents from host tissues. Acting both in concert and individually, the diverse cells comprising the epithelial layer play the functions of protection (1), sensation (2, 3), transport of substances (4, 5), and repair (6). Colonic epithelial cells belong to three lineages: Absorptive enterocytes, enteroendocrine cells, and goblet cells (GCs). The colonic epithelium is morphologically organized into repeating units called crypts of Lieberkühn, where stem cells located at the base of the crypts divide and successively differentiate into the mature lineages as they migrate toward the crypt surface (7). Many of the key specialized functions of epithelial cells are, in part, defined by proteins involved in ion transport, located either on their luminal or basolateral membrane. Thus, among different gastrointestinal epithelial cells, ion channels, carriers, exchangers, and pumps work in concert to define a variety of essential functions: 1) Solute and electrolyte absorption and secretion in absorptive enterocytes (reviewed in refs. 5 and 8); 2) environment sensation and serotonin secretion by enteroendocrine cells (2, 9); and 3) mucus secretion by GCs and subsequent mucus maturation into the protective layer covering the epithelial surface (1012). Despite this progress, ionic transport in GCs and its implications in GC physiology is a topic that remains poorly understood. Here, we address the role of the Ca2+- and voltage-activated K+ channel (BK channel) in GCs.GCs play two primary roles: One related to the maintenance of the mucosal barrier (reviewed in refs. 1 and 13) and one related with the mucosal immune homeostasis (reviewed in refs. 14 and 15). The role of GCs in barrier maintenance consists in generation of the mucus layer lining the intestinal lumen. One way GCs carry out this role is by secreting MUC2, the gel-forming mucin that forms the scaffold of the mucus layer separating luminal pathogens and commensal microbiota from the epithelial surface (11, 12, 15, 16). This separation is critical, as has been demonstrated in both animal models and humans: Mouse models with deficient mucus layer generation develop spontaneous colitis (16, 17), whereas a more penetrable mucus layer has been observed in patients with ulcerative colitis (UC), a form of human inflammatory bowel disease (IBD) (18, 19). The constant replenishment of the mucus layer involves MUC2 exocytosis from GCs, and subsequent maturation (hydration and expansion) of the secreted MUC2 to form the gel-like mucus coating the epithelium (15). Both exocytosis and maturation of MUC2 are highly dependent on anion and K+ transport (1012, 20). It has been proposed that mucin exocytosis in colon requires activities of the Na+/K+/2Cl cotransporter (NKCC1) (20, 21), and also anion and K+ channels whose identities are still unclear (20). It is also not clearly known whether specific ionic conductances are intrinsic to GCs or are located in the surrounding absorptive enterocytes. Although several types of K+ channels—including KCa3.1, Kv7.1, and BK channels—have been found in colonic epithelial cells (2227), to what extent any of those K+ channels are specifically associated with GCs or critical to their function remains unclear. To date, most functional studies about colonic K+ channels have focused on their roles in electrolyte and fluid secretion/absorption of the whole colon, whereas the cellular events relating K+ channels to specific roles in GC function are still poorly understood.Among colonic epithelial K+ channels, the BK channel (also known as KCa1.1), the Ca2+- and voltage-activated K+ channel of high conductance, has been proposed to be the main component of colonic K+ secretion into the lumen (2830). BK channels are homotetramers of the pore-forming BKα subunit, but can also contain tissue-specific regulatory subunits that critically define the functional properties of the channel (31). BK channels composed exclusively of the pore-forming BKα subunit are unlikely to be activated at the physiological conditions of epithelial cells and, as a consequence, the molecular properties of colonic BK channels that would allow them to contribute to colonic ion transport remain unclear. Recently, we established that the leucine-rich repeat-containing protein 26 (LRRC26), a BK regulatory γ-subunit, is specifically expressed in secretory epithelial cells, including GCs of the gastrointestinal tract (32). When LRRC26 is present in a BK channel complex, the resulting channel activates near normal resting physiological conditions, even in the absence of any elevation of intracellular Ca2+ (33).In the present study, we have specifically probed the role of BK channels in cells of the colonic epithelium and examined the impact of deletions of either the BKα subunit or LRRC26 on colonic function. Here, through recordings from identified GCs in intact colonic crypts, we show that LRRC26-associated BK channels contribute the major K+ current at low intracellular Ca2+ (∼250 nM) in mouse colonic GCs. Furthermore, the LRRC26-containing BK channels are activated near −40 mV, even in the absence of intracellular Ca2+. In contrast, in identified GCs from Lrrc26−/− mice, BK current is present, but it is only activated at membrane potentials unlikely to ever occur physiologically. Surprisingly, all colonic epithelial MUC2 cells sampled completely lack functional BK channels. To establish that the LRRC26-containing BK channels contribute to normal K+ fluxes in intact colon tissue, we show that the transepithelial current across distal colon at rest has a component dependent on LRRC26-associated BK channels, which is absent when either BKα or LRRC26 is genetically deleted. Moreover, the genetic ablation of either LRRC26 or BK channel results in a dramatically enhanced susceptibility to colitis induced by dextran sodium sulfate (DSS). Overall, our results suggest that normal potassium flux through LRRC26-associated BK channels in GCs has a protective role against development of colitis.  相似文献   

6.
Neuropeptides and neurotrophic factors secreted from dense core vesicles (DCVs) control many brain functions, but the calcium sensors that trigger their secretion remain unknown. Here, we show that in mouse hippocampal neurons, DCV fusion is strongly and equally reduced in synaptotagmin-1 (Syt1)- or Syt7-deficient neurons, but combined Syt1/Syt7 deficiency did not reduce fusion further. Cross-rescue, expression of Syt1 in Syt7-deficient neurons, or vice versa, completely restored fusion. Hence, both sensors are rate limiting, operating in a single pathway. Overexpression of either sensor in wild-type neurons confirmed this and increased fusion. Syt1 traveled with DCVs and was present on fusing DCVs, but Syt7 supported fusion largely from other locations. Finally, the duration of single DCV fusion events was reduced in Syt1-deficient but not Syt7-deficient neurons. In conclusion, two functionally redundant calcium sensors drive neuromodulator secretion in an expression-dependent manner. In addition, Syt1 has a unique role in regulating fusion pore duration.

To date, over 100 genes encoding neuropeptides and neurotrophic factors, together referred to as neuromodulators, are identified, and most neurons express neuromodulators and neuromodulator receptors (1). Neuromodulators travel through neurons in dense core vesicles (DCVs) and, upon secretion, regulate neuronal excitability, synaptic plasticity, and neurite outgrowth (24). Dysregulation of DCV secretion is linked to many brain disorders (57). However, the molecular mechanisms that regulate neuromodulator secretion remain largely elusive.Neuromodulator secretion, like neurotransmitter secretion from synaptic vesicles (SVs), is tightly controlled by Ca2+. The Ca2+ sensors that regulate secretion have been described for other secretory pathways but not for DCV exocytosis in neurons. Synaptotagmin (Syt) and Doc2a/b are good candidate sensors due to their interaction with SNARE complexes, phospholipids, and Ca2+ (811). The Syt family consists of 17 paralogs (12, 13). Eight show Ca2+-dependent lipid binding: Syt1 to 3, Syt5 to 7, and Syt9 and 10 (14, 15). Syt1 mediates synchronous SV fusion (8), consistent with its low Ca2+-dependent lipid affinity (15, 16) and fast Ca2+/membrane dissociation kinetics (16, 17). Syt1 is also required for the fast fusion in chromaffin cells (18) and fast striatal dopamine release (19). Synaptotagmin-7 (Syt7), in contrast, drives asynchronous SV fusion (20), in line with its a higher Ca2+ affinity (15) and slower dissociation kinetics (16). Syt7 is also a major calcium sensor for neuroendocrine secretion (21) and secretion in pancreatic cells (2224). Other sensors include Syt4, which negatively regulates brain-derived neurothropic factor (25) and oxytocin release (26), in line with its Ca2+ independency. Syt9 regulates hormone secretion in the anterior pituitary (27) and, together with Syt1, secretion from PC12 cells (28, 29). Syt10 controls growth factor secretion (30). However, Syt9 and Syt10 expression is highly restricted in the brain (3133). Hence, the calcium sensors for neuronal DCV fusion remain largely elusive. Because DCVs are generally not located close to Ca2+ channels (34), we hypothesized that DCV fusion is triggered by high-affinity Ca2+ sensors. Because of their important roles in vesicle secretion, their Ca2+ binding ability, and their high expression levels in the brain (20, 31, 3538), we addressed the roles of Doc2a/b, Syt1, and Syt7 in neuronal DCV fusion.In this study, we used primary Doc2a/b-, Syt1-, and Syt7-null (knockout, KO) neurons expressing DCV fusion reporters (34, 3941) with single-vesicle resolution. We show that both Syt1 and Syt7, but not Doc2a/b, are required for ∼60 to 90% of DCV fusion events. Deficiency of both Syt1 and Syt7 did not produce an additive effect, suggesting they function in the same pathway. Syt1 overexpression (Syt1-OE) rescued DCV fusion in Syt7-null neurons, and vice versa, indicating that the two proteins compensate for each other in DCV secretion. Moreover, overexpression of Syt1 or Syt7 in wild-type (WT) neurons increased DCV fusion, suggesting they are both rate limiting for DCV secretion. We conclude that DCV fusion requires two calcium sensors, Syt1 and Syt7, that act in a single/serial pathway and that both sensors regulate fusion in a rate-limiting and dose-dependent manner.  相似文献   

7.
8.
Cerebellar granule cells (GrCs) are usually regarded as a uniform cell type that collectively expands the coding space of the cerebellum by integrating diverse combinations of mossy fiber inputs. Accordingly, stable molecularly or physiologically defined GrC subtypes within a single cerebellar region have not been reported. The only known cellular property that distinguishes otherwise homogeneous GrCs is the correspondence between GrC birth timing and the depth of the molecular layer to which their axons project. To determine the role birth timing plays in GrC wiring and function, we developed genetic strategies to access early- and late-born GrCs. We initiated retrograde monosynaptic rabies virus tracing from control (birth timing unrestricted), early-born, and late-born GrCs, revealing the different patterns of mossy fiber input to GrCs in vermis lobule 6 and simplex, as well as to early- and late-born GrCs of vermis lobule 6: sensory and motor nuclei provide more input to early-born GrCs, while basal pontine and cerebellar nuclei provide more input to late-born GrCs. In vivo multidepth two-photon Ca2+ imaging of axons of early- and late-born GrCs revealed representations of diverse task variables and stimuli by both populations, with modest differences in the proportions encoding movement, reward anticipation, and reward consumption. Our results suggest neither organized parallel processing nor completely random organization of mossy fiber→GrC circuitry but instead a moderate influence of birth timing on GrC wiring and encoding. Our imaging data also provide evidence that GrCs can represent generalized responses to aversive stimuli, in addition to recently described reward representations.

Cerebellar granule cells (GrCs) comprise the majority of neurons in the mammalian brain (1, 2). Each GrC receives only four excitatory inputs from mossy fibers, which originate in a variety of brainstem nuclei and the spinal cord, and the vast number of GrCs permits diverse combinations of mossy fiber inputs. Classical theories of cerebellar function have therefore proposed that GrCs integrate diverse, multimodal mossy fiber inputs and thus collectively expand coding space in the cerebellum (35). Until recently, studies have focused on the role of GrCs in implementing sparse coding of sensorimotor variables and stimuli (69). However, recent physiological studies of GrCs in awake, behaving animals highlight GrC encoding of cognitive signals in addition to sensorimotor signals (1013). GrCs have also been recently shown to encode denser representations than expected by classical theory (1012, 1418), including a lack of dimensionality expansion under certain conditions (18).Despite the vast number of GrCs, stable molecularly or physiologically defined GrC subtypes within a single cerebellar region or lobule have not been described (1922), although variation in gene expression across different regions has been reported (22, 23). The only known axis along which spatially intermingled GrCs can be distinguished from each other is the depth of the molecular layer to which their parallel fiber axons (PFs) project, which is dictated by GrC lineage and birth timing (24, 25). Birth timing predicts the wiring and functional properties of diverse neuron types in many neural systems (26), including the neocortex (27, 28), other forebrain regions (29, 30), olfactory bulb (3133), and ventral spinal cord (34, 35). Furthermore, classic studies utilizing γ-irradiation at different times during rat postnatal development to ablate different cerebellar GrC and interneuron populations suggested that GrCs born at different times could contribute differentially to motor vs. action coordination (36). These observations also led to an as-of-yet untested hypothesis that mossy fibers arriving at different times during development could connect with different GrC populations. Could GrC birth timing be an organizing principle for information processing in the cerebellum?Recent evidence and modeling point to the possibility of spatial clusters of coactivated PFs (15, 37), suggesting that GrCs born around the same time may disproportionally receive coactive mossy fiber inputs. However, another study using different methods and stimuli did not find differences in the physiological responses of early- and late-born GrCs to various sensorimotor stimuli (38). Here, we address the role of birth timing in GrC wiring and function. We developed strategies to gain genetic access to early- and late-born GrCs, as well as control GrCs not restricted by birth timing. We report the first monosynaptic input tracing to GrCs, finding differential mossy fiber inputs to GrCs in vermis lobule 6 and simplex, as well as different patterns of input to early- and late-born GrCs in vermis lobule 6. Finally, we performed in vivo multidepth two-photon Ca2+ imaging of PFs of early- and late-born GrCs during an operant task and presentation of a panel of sensory, appetitive, and aversive stimuli. We found modest differences in the proportions of early- and late-born GrCs encoding of a subset of movement and reward parameters. Together, these results reveal a contribution of GrC birth timing to their input wiring and diverse encoding properties.  相似文献   

9.
Organic Fenton-like catalysis has been recently developed for water purification, but redox-active compounds have to be ex situ added as oxidant activators, causing secondary pollution problem. Electrochemical oxidation is widely used for pollutant degradation, but suffers from severe electrode fouling caused by high-resistance polymeric intermediates. Herein, we develop an in situ organic Fenton-like catalysis by using the redox-active polymeric intermediates, e.g., benzoquinone, hydroquinone, and quinhydrone, generated in electrochemical pollutant oxidation as H2O2 activators. By taking phenol as a target pollutant, we demonstrate that the in situ organic Fenton-like catalysis not only improves pollutant degradation, but also refreshes working electrode with a better catalytic stability. Both 1O2 nonradical and ·OH radical are generated in the anodic phenol conversion in the in situ organic Fenton-like catalysis. Our findings might provide a new opportunity to develop a simple, efficient, and cost-effective strategy for electrochemical water purification.

The efficient generation of reactive oxygen species is essential for pollutant degradation in water purification. The metal-mediated Fenton catalysis has been widely used for several decades owning to its high efficiency, low cost, and easy operation (1). However, it has several technical drawbacks to largely limit further applications, e.g., harsh pH, metal-rich sludge, secondary pollution, and poor stability (1). Alternatively, the metal-free Fenton catalysis has recently attracted increasing interests. Redox-active compounds serve as the oxidant activator to decompose pollutants via radical and/or nonradical pathways (25). These pathways depend highly on the atomic and electronic structures and molecular configurations of compounds and their molecular interactions with oxidants (618). So far organic activators are ex situ introduced and cause secondary pollution, although the performance can be largely improved (218). Such an intrinsic drawback greatly restricts its practical applications. Thus, in situ organic Fenton-like catalysis without secondary pollution is greatly desired for clean and safe water purification.Electrochemical oxidation (EO) at low bias is widely used for pollutant degradation owning to its high current efficiency and low energy consumption, but largely suffers from electrode fouling (19, 20). Such fouling is mainly caused by anodic polymeric intermediates with large molecular size, low geometric polarity, and high structural stability, thus anodic oxidation is thermodynamically terminated at this stage (19, 20). How to remove polymeric intermediates is essential for electrochemical water purification. It is interesting to note that anodic polymeric intermediates usually contain quinonelike moieties (C = O) and persistent organic radicals, as the electrons in nucleophilic C-OH can be readily transferred to generate C-O· and C = O (19, 20). Quinonelike moieties are redox-active because of their high electron density and strong electron-donating properties, thus can serve as the metal ligand and reductant to enhance transition-metal redox cycling, and also be involved in the environmental geochemistry of natural organic matters (2130). Moreover, quinonelike moieties and persistent organic radicals can directly serve as an organic activator to initiate organic Fenton-like catalysis for environmental remediation (3140). Thus, these redox-active anodic polymeric intermediates are likely to trigger organic Fenton-like catalysis.Inspired by above analyses, we constructed and validated in situ organic Fenton-like catalysis for electrochemical water purification at low bias before oxygen evolution (Scheme 1). Phenol, a model chemical widely present in environments, and other typical halogenated and nonhalogenated aromatic compounds were selected as target pollutants. Carbon felt (CF), a model material with high activity and low cost, and other typical dimensionally stable anodes were selected as target electrodes. Reaction systems were named in the form of “EO + ex situ added reagent + cathode,” as their anodes were identical. Pollutant degradation and electrode antifouling performances were evaluated under various conditions. After the major reactive oxygen species were identified using a suite of testing methods, and the potential role of trace transition metals, especially iron and copper, was examined, the possible molecular mechanism of the in situ organic Fenton-like catalysis was proposed.Open in a separate windowScheme 1.Scheme diagrams of the EO-Ti, EO/H2O2-Ti, and EO/O2-CF systems.  相似文献   

10.
Calcium (Ca2+)-dependent protein kinases (CDPKs or CPKs) are a unique family of Ca2+ sensor/kinase-effector proteins with diverse functions in plants. In Arabidopsis thaliana, CPK28 contributes to immune homeostasis by promoting degradation of the key immune signaling receptor-like cytoplasmic kinase BOTRYTIS-INDUCED KINASE 1 (BIK1) and additionally functions in vegetative-to-reproductive stage transition. How CPK28 controls these seemingly disparate pathways is unknown. Here, we identify a single phosphorylation site in the kinase domain of CPK28 (Ser318) that is differentially required for its function in immune homeostasis and stem elongation. We show that CPK28 undergoes intermolecular autophosphorylation on Ser318 and can additionally be transphosphorylated on this residue by BIK1. Analysis of several other phosphorylation sites demonstrates that Ser318 phosphorylation is uniquely required to prime CPK28 for Ca2+ activation at physiological concentrations of Ca2+, possibly through stabilization of the Ca2+-bound active state as indicated by intrinsic fluorescence experiments. Together, our data indicate that phosphorylation of Ser318 is required for the activation of CPK28 at low intracellular [Ca2+] to prevent initiation of an immune response in the absence of infection. By comparison, phosphorylation of Ser318 is not required for stem elongation, indicating pathway-specific requirements for phosphorylation-based Ca2+-sensitivity priming. We additionally provide evidence for a conserved function for Ser318 phosphorylation in related group IV CDPKs, which holds promise for biotechnological applications by generating CDPK alleles that enhance resistance to microbial pathogens without consequences to yield.

Protein kinases represent one of the largest eukaryotic protein superfamilies. While roughly 500 protein kinases have been identified in humans (1), the genomes of Arabidopsis thaliana (hereafter, Arabidopsis) (2) and Oryza sativa (3) encode more than 1,000 and 1,500 protein kinases, respectively, including several families unique to plants. Among these protein kinases are the receptor-like kinases (RLKs), receptor-like cytoplasmic kinases (RLCKs), and calcium-dependent protein kinases (CDPKs or CPKs) that have emerged as key regulators of plant immunity (46). Despite encompassing only 2% of most eukaryotic genomes, protein kinases phosphorylate more than 40% of cellular proteins (7, 8), reflecting their diverse roles in coordinating intracellular signaling events. Reversible phosphorylation of serine (Ser), threonine (Thr), and tyrosine (Tyr) residues can serve an array of functions including changes in protein conformation and activation state (9, 10), protein stability and degradation (11, 12), subcellular localization (1315), and interaction with protein substrates (1618).Calcium (Ca2+) is a ubiquitous secondary messenger that acts cooperatively with protein phosphorylation to propagate intracellular signals. Spatial and temporal changes in intracellular Ca2+ levels occur in response to environmental and developmental cues (1923). In plants, Ca2+ transients are decoded by four major groups of calcium sensor proteins, which possess one or more Ca2+-binding EF-hand motifs (24, 25): calmodulins (CaM), CaM-like proteins, calcineurin B–like proteins, CDPKs, and Ca2+/CaM-dependent protein kinases.At the intersection of phosphorylation cascades and Ca2+ signaling are CDPKs, a unique family of Ca2+ sensor/kinase-effector proteins. CDPKs have been identified in all land plants and green algae, as well as certain protozoan ciliates and apicomplexan parasites (26, 27). CDPKs have a conserved domain architecture, consisting of a canonical Ser/Thr protein kinase domain and an EF-hand containing Ca2+-binding CaM-like domain (CLD), linked together by an autoinhibitory junction (AIJ) and flanked by variable regions on both the amino (N) and carboxyl (C) termini (28, 29). As their name implies, most CDPKs require Ca2+ for their activation (30). Upon Ca2+ binding to all EF-hands in the CaM-like domain, a dramatic conformational change occurs, freeing the AIJ from the catalytic site of the kinase, rendering the enzyme active (3133). CDPKs vary in their sensitivity to Ca2+ (30), presumably allowing proteins to perceive distinct stimuli through differences in Ca2+-binding affinity. For example, Arabidopsis CPK4 displays half maximal kinase activity in the presence of ∼3 μM free Ca2+ (30) while CPK5 only requires ∼100 nM (34). Importantly, CDPKs are signaling hubs with documented roles in multiple distinct pathways (4, 24, 3538) and are therefore likely regulated beyond Ca2+ activation.Subfunctionalization is at least partially mediated by protein localization and interaction with pathway-specific binding partners, as is well documented for Arabidopsis CPK3 which functions in response to biotic and abiotic stimuli in distinct cellular compartments (39). Recent attention has been drawn to site-specific phosphorylation as a mechanism to regulate the activity of multifunctional kinases. For example, phosphorylation sites on the RLK BRASSINOSTEROID INSENSITIVE 1–ASSOCIATED KINASE 1 (BAK1) are differentially required for its function as a coreceptor with a subset of leucine-rich repeat –RLKs (40). Phosphoproteomic analyses indicate that CDPKs are differentially phosphorylated following exposure to distinct stimuli (4148); however, the biochemical mechanisms by which site-specific phosphorylation regulates multifunctional CDPKs is still poorly understood.Arabidopsis CPK28 is a plasma membrane–localized protein kinase with dual roles in plant immune homeostasis (4951) and phytohormone-mediated reproductive growth (52, 53). In vegetative plants, CPK28 serves as a negative regulator of immune signal amplitude by phosphorylating and activating two PLANT U-BOX–type E3 ubiquitin ligases, PUB25 and PUB26, which target the key immune RLCK BOTRYTIS-INDUCED KINASE 1 (BIK1) for proteasomal degradation (50). Owing to elevated levels of BIK1, CPK28 null plants (cpk28-1) have heightened immune responses and enhanced resistance to the bacterial pathogen Pseudomonas syringae pv. tomato DC3000 (Pto DC3000) (51). Upon transition to the reproductive stage, cpk28-1 plants additionally present shorter leaf petioles, enhanced anthocyanin production, and a reduction in stem elongation (52, 53). The molecular basis for developmental phenotypes in the cpk28-1 knockout mutant, beyond hormonal imbalance (52, 53), are comparatively unknown.Our recent work demonstrated that autophosphorylation status dictates Ca2+-sensitivity of CPK28 peptide kinase activity in vitro (54). While dephosphorylated CPK28 is stimulated by the addition of 100 μM CaCl2 compared to untreated protein, hyperphosphorylated CPK28 displayed similar levels of activity at basal Ca2+ concentrations (54). These results highlight the interesting possibility that phosphorylation status may control the activation of multifunctional kinases in distinct pathways by allowing CDPKs to respond to stimulus-specific Ca2+ signatures.In the present study, we identify a single autophosphorylation site, Ser318, that decouples the activity of CPK28 in immune signaling from its role in reproductive growth. We show that expression of a nonphosphorylatable Ser-to-Ala variant (CPK28S318A) is unable to complement the immune phenotypes of cpk28-1 mutants but is able to complement defects in stem growth. Further, we uncover a functional role for phosphorylation of Ser318 in priming CPK28 for activation at low free [Ca2+]. Together, we demonstrate that site-specific phosphorylation can direct the activity of a multifunctional kinase in distinct pathways and provide evidence for a conserved mechanism in orthologous group IV CDPKs.  相似文献   

11.
Mitochondrial ATP production is a well-known regulator of neuronal excitability. The reciprocal influence of plasma-membrane potential on ATP production, however, remains poorly understood. Here, we describe a mechanism by which depolarized neurons elevate the somatic ATP/ADP ratio in Drosophila glutamatergic neurons. We show that depolarization increased phospholipase-Cβ (PLC-β) activity by promoting the association of the enzyme with its phosphoinositide substrate. Augmented PLC-β activity led to greater release of endoplasmic reticulum Ca2+ via the inositol trisphosphate receptor (IP3R), increased mitochondrial Ca2+ uptake, and promoted ATP synthesis. Perturbations that decoupled membrane potential from this mode of ATP synthesis led to untrammeled PLC-β–IP3R activation and a dramatic shortening of Drosophila lifespan. Upon investigating the underlying mechanisms, we found that increased sequestration of Ca2+ into endolysosomes was an intermediary in the regulation of lifespan by IP3Rs. Manipulations that either lowered PLC-β/IP3R abundance or attenuated endolysosomal Ca2+ overload restored animal longevity. Collectively, our findings demonstrate that depolarization-dependent regulation of PLC-β–IP3R signaling is required for modulation of the ATP/ADP ratio in healthy glutamatergic neurons, whereas hyperactivation of this axis in chronically depolarized glutamatergic neurons shortens animal lifespan by promoting endolysosomal Ca2+ overload.

Spatially circumscribed ATP production at nerve termini is predicated on local mitochondria that are energized when voltage-gated Ca2+ channels provide the [Ca2+] elevations needed to overcome the low sensitivity of the mitochondrial Ca2+ uniporter (MCU) (13). In neuronal soma, however, bulk cytosolic [Ca2+] is not elevated to levels needed for mitochondrial sequestration. Rather, mitochondrial Ca2+ uptake in the somatodendritic compartment occurs at specialized points of contact between mitochondria and endoplasmic reticulum (ER) where Ca2+ released by IP3Rs is transferred into the mitochondrial matrix (4). Approximately 75 to 90% of the somatic ATP synthesized following interorganellar transfer of Ca2+ is consumed by Na+/K+ ATPases, which help establish resting membrane potential and permit repolarization during activity (5, 6). Therefore, defects in neuronal ATP synthesis result in loss of membrane potential and hyperexcitability (6).Whether excitability of the somatic plasma membrane (PM) exerts reciprocal influence on mitochondrial [Ca2+] and ATP production remains poorly understood. In an attempt to fill some of the gaps in knowledge, we examined the effects of PM potential on mitochondrial ATP production and Ca2+ homeostasis in Drosophila neurons. Owing to recent reports of neuronal hyperexcitability being a driver of diminished longevity in organisms ranging from Caenorhabditis elegans to humans (79), we hoped our studies would inform insights into the regulation of aging and lifespan. Moreover, since neuronal hyperexcitability, Ca2+ dyshomeostasis, and bioenergetic dysfunction characterize neurodegenerative diseases (6, 10, 11), uncovering actionable molecular targets that bridge these perturbations may bear therapeutic value. Our findings reveal a previously unknown mechanism by which excitability regulates bioenergetics and Ca2+ signaling and points to the utility of this signaling circuit in the regulation of longevity.  相似文献   

12.
Priming of CD8+ T cells by dendritic cells (DCs) is crucial for the generation of effective antitumor immune responses. Here, we describe a liposomal vaccine carrier that delivers tumor antigens to human CD169/Siglec-1+ antigen-presenting cells using gangliosides as targeting ligands. Ganglioside-liposomes specifically bound to CD169 and were internalized by in vitro-generated monocyte-derived DCs (moDCs) and macrophages and by ex vivo-isolated splenic macrophages in a CD169-dependent manner. In blood, high-dimensional reduction analysis revealed that ganglioside-liposomes specifically targeted CD14+ CD169+ monocytes and Axl+ CD169+ DCs. Liposomal codelivery of tumor antigen and Toll-like receptor ligand to CD169+ moDCs and Axl+ CD169+ DCs led to cytokine production and robust cross-presentation and activation of tumor antigen-specific CD8+ T cells. Finally, Axl+ CD169+ DCs were present in cancer patients and efficiently captured ganglioside-liposomes. Our findings demonstrate a nanovaccine platform targeting CD169+ DCs to drive antitumor T cell responses.

The major breakthrough of immune-checkpoint inhibitors, such as anti-CTLA4 and anti–PD-L1, in cancer therapy is still limited to a minority of patients who respond to this treatment (1). Patients with pancreatic cancer, for example, failed to respond to monotherapies of checkpoint inhibitors in multiple trials (2, 3). Factors such as poor tumor immunogenicity, tumor-immunosuppressive microenvironment, and the lack of an existing tumor-specific immune response are thought to contribute to patients’ lack of response to these immune-checkpoint inhibitors (2, 4, 5). Nevertheless, the abundance of intratumoral CD8+ T cells is associated with longer survival of pancreatic cancer patients, suggesting these patients may benefit from a better antitumor immunity (68). Therefore, new strategies aiming to boost patients’ antitumor CD8+ T cell responses should be explored to improve current therapies.Dendritic cells (DCs) play a crucial role in eliciting immune responses against tumor-specific antigens and have therefore generated significant interest as a therapeutic target in the context of cancer immunotherapy (9). The most commonly used DC-based immunotherapy utilizes monocyte-derived DCs (moDCs) due to the large numbers that can be generated ex vivo. In general, moDC-based vaccines have shown some survival benefit and appear to be well-tolerated; however, the objective response rate in most studies is still relatively low (9, 10). Moreover, since generating DCs ex vivo is a laborious, time-consuming, and costly process, research is shifting toward targeting tumor antigens to naturally circulating or tissue-resident DCs in vivo as a vaccine strategy to induce immune responses (11). Both in mice and humans, DCs can be divided into several subsets, of which the conventional DCs (CD141+ cDC1 and CD1c+ cDC2) have been shown to be responsible for T cell priming (12, 13).In vivo DC targeting can be achieved by using antibodies or ligands that bind to DC-specific receptors and are directly conjugated to tumor antigen or to nanoparticles harboring tumor antigen. Targeting C-type lectin receptors in particular, such as DEC-205, Clec-9A, and DC-SIGN, has been demonstrated to induce antigen-specific and antitumor responses in mouse and human models (1417). Recently, we compared two vaccination strategies of antigen–antibody conjugates directed to either DEC-205+ DCs or to CD169+ macrophages, a type of macrophage that acts as sentinel in secondary lymphoid organs (18). Remarkably, we observed that antigen targeting toward CD169+ macrophages led to a significant antigen-specific CD8+ T cell response that was as efficient as DEC-205 targeting and capable of suppressing tumor cell outgrowth (1820). Stimulation of antigen-specific immune responses by targeting to CD169 has also been demonstrated using HLA-A2.1 transgenic mice and human CD169-expressing moDCs (21), indicating the immunotherapy potential of antigen targeting to CD169.In a resting state, CD169/Siglec-1 is highly expressed by a specific subtype of macrophages that are located bordering the marginal zone in the spleen and the subcapsular sinus of lymph nodes (22, 23). Their strategic location allows them to be among the first cells to encounter and to capture blood and lymph-borne pathogens, and, in conjunction with DCs, to initiate the appropriate immune responses (18, 19, 24, 25). In addition to combating infection, CD169+ macrophages have been implicated in antitumor immunity. They have been shown to capture tumor-derived materials in mouse and human (26, 27), and their frequency in tumor-draining lymph nodes is clearly associated with better clinical outcomes in several types of cancer (2830). Although the exact mechanism is unclear, these observations suggest that lymphoid-resident CD169+ macrophages can positively contribute to antitumor immunity. Next to lymphoid tissue-resident macrophages, CD169 is also constitutively expressed by a recently described Axl+ Siglec6+ DC subset (Axl+ DCs, AS DCs, or pre-DCs) present in peripheral blood and lymphoid tissues (3134). Axl+ DCs have been proposed as a distinct DC subset that has the capacity to produce inflammatory cytokines and to stimulate CD4+ and CD8+ T cells (3133). In addition to these constitutively CD169-expressing macrophages and DCs, during inflammatory conditions, monocytes can up-regulate CD169 in response to type I interferons (IFN-Is) (35, 36).CD169 is a member of the sialic acid-binding Ig-like lectin (Siglec) receptor family that recognizes sialic acids present on glycoproteins or glycolipids on the cell surface and mediates cell–cell interactions and adhesion (37). Sialic acid-containing glycosphingolipids, such as GM3, GT1b, and GD1a gangliosides, are known to be endogenous ligands for CD169 molecules (38, 39). However, the CD169–sialic acid axis can be hijacked as a receptor entry molecule by viral pathogens, including murine leukemia virus (MLV), HIV, and Ebola virus to infect DCs or macrophages (4043). The CD169-mediated entry and transinfection is dependent on gangliosides, including GM3, that are present on the viral lipid membrane (40, 44, 45). Interestingly, Axl+ DCs have been recently demonstrated to be the predominant DC subset to capture HIV in a CD169-dependent manner.In this study, we aimed to exploit ganglioside–CD169 interactions to develop a novel tumor antigen vaccination strategy that directs tumor antigens to human CD169+ antigen-presenting cells (APCs) using liposomes containing gangliosides. We generated liposomes with different types of gangliosides and assessed the binding and uptake by different types of human CD169+ APCs, including monocytes and primary and monocyte-derived macrophages and DCs. High-dimensionality mapping revealed the specificity of ganglioside-liposome targeting exclusively to circulating CD169+ monocytes and Axl+ DCs. To determine the efficacy of ganglioside-liposomes for antigen presentation, we encapsulated peptides derived from the pancreatic cancer-associated tumor antigen Wilms tumor 1 (WT1) or melanoma-associated gp100 antigen into the ganglioside-liposomes. CD169+ moDCs and Axl+ DCs loaded with these ganglioside-liposomes efficiently activated CD8+ T cells specific for these epitopes. Moreover, Axl+ DCs were present in patients with four different cancers and could be targeted by ganglioside-liposomes. Our data demonstrate that ganglioside-liposomes can be used as nanovaccine carriers that efficiently target CD169+ DCs for cross-presentation and antigen-specific T cell activation. In conclusion, our studies support the concept that cancer vaccines targeting to CD169 can be applied to boost CD8+ T cell responses in cancer patients.  相似文献   

13.
The cyclic adenosine monophosphate (cAMP)-dependent potentiation of neurotransmitter release is important for higher brain functions such as learning and memory. To reveal the underlying mechanisms, we applied paired pre- and postsynaptic recordings from hippocampal mossy fiber-CA3 synapses. Ca2+ uncaging experiments did not reveal changes in the intracellular Ca2+ sensitivity for transmitter release by cAMP, but suggested an increase in the local Ca2+ concentration at the release site, which was much lower than that of other synapses before potentiation. Total internal reflection fluorescence (TIRF) microscopy indicated a clear increase in the local Ca2+ concentration at the release site within 5 to 10 min, suggesting that the increase in local Ca2+ is explained by the simple mechanism of rapid Ca2+ channel accumulation. Consistently, two-dimensional time-gated stimulated emission depletion microscopy (gSTED) microscopy showed an increase in the P/Q-type Ca2+ channel cluster size near the release sites. Taken together, this study suggests a potential mechanism for the cAMP-dependent increase in transmission at hippocampal mossy fiber-CA3 synapses, namely an accumulation of active zone Ca2+ channels.

Communication between neurons is largely mediated by chemical synapses. Synaptic strengths are not fixed, but change dynamically in the short and longer term in an activity-dependent manner (short- and long-term plasticity, 13). Moreover, neuromodulators act on presynaptic terminals to modulate synaptic strength. Such activity-dependent or modulatory changes are often mediated by the activation of second messengers, such as protein kinase A and C (2). Second messenger systems, particularly the cyclic adenosine monophosphate (cAMP)/PKA-dependent system, are important for higher brain functions, including learning and memory in Aplysia (3), flies (4, 5), and the mammalian brain (6). Despite its functional importance, the cellular and molecular mechanisms of cAMP-dependent modulation are still poorly understood regardless of whether Aplysia synapses and Drosophila neuromuscular junctions have been investigated (2, 7). Mammalian central synapses are no exception here, also reflecting technical difficulties due to the generally small size of the presynaptic terminals in the mammalian brain.Hippocampal mossy fiber-CA3 (MF-CA3) synapses are characterized by exceptionally large presynaptic terminals (hippocampal mossy fiber bouton, hMFB), which allow for the direct analysis of the cellular mechanisms of synaptic transmission and plasticity by using patch-clamp recordings (810). Thus, hMFBs provide a suitable model of cortical synapses in the mammalian brain. Moreover, these synapses are functionally important for brain function such as pattern separation (11). Mossy fiber synapses are known to exhibit unique presynaptic forms of short- and long-term synaptic potentiation and depression, which share the cAMP/PKA-dependent induction mechanism (1215). In addition, the cAMP-dependent plasticity pathway is important for presynaptic modulation by dopamine and noradrenaline (1618), which modulates hippocampal network activity and behavior. However, its underlying cellular mechanisms remain largely unclear. Enhancement of the molecular priming and docking of synaptic vesicles at mossy fiber synapses has been suggested by previous studies using genetics and electron microscopy (1921). In particular, RIM1, an active zone scaffold protein, is crucial for cAMP-dependent long-term potentiation (LTP) (19) and is phosphorylated by PKA, although a corresponding phosphorylation mutant of RIM1 was found to have no effect on long-term potentiation (22, but see ref. 23). Other studies on hMFBs have implicated a role in positional priming, i.e., changes in the spatial coupling between Ca2+ channels and the release machinery (24). However, there is a lack of the direct visualization or manipulation of this regulation.In order to measure the intracellular Ca2+ sensitivity of transmitter release directly and examine the mechanisms of cAMP-dependent modulation quantitatively, we here carried out Ca2+ uncaging experiments at hippocampal mossy fiber synapses. Unexpectedly, our results failed to show changes in Ca2+ sensitivity, but instead uncovered an increase in local Ca2+ concentrations at the release sites. Furthermore, by live imaging of local Ca2+ using total internal reflection fluorescence (TIRF) microscopy as well as superresolution time gated STED (gSTED) microscopy, we provided evidence that rather rapid Ca2+ channel accumulation may underlie cAMP-induced potentiation instead of release machinery modulations. This study thus provides a potential mechanism of presynaptic modulation at central synapses.  相似文献   

14.
NKCC1 is the primary transporter mediating chloride uptake in immature principal neurons, but its role in the development of in vivo network dynamics and cognitive abilities remains unknown. Here, we address the function of NKCC1 in developing mice using electrophysiological, optical, and behavioral approaches. We report that NKCC1 deletion from telencephalic glutamatergic neurons decreases in vitro excitatory actions of γ-aminobutyric acid (GABA) and impairs neuronal synchrony in neonatal hippocampal brain slices. In vivo, it has a minor impact on correlated spontaneous activity in the hippocampus and does not affect network activity in the intact visual cortex. Moreover, long-term effects of the developmental NKCC1 deletion on synaptic maturation, network dynamics, and behavioral performance are subtle. Our data reveal a neural network function of NKCC1 in hippocampal glutamatergic neurons in vivo, but challenge the hypothesis that NKCC1 is essential for major aspects of hippocampal development.

Intracellular chloride concentration ([Cl]i) is a major determinant of neuronal excitability, as synaptic inhibition is primarily mediated by chloride-permeable receptors (1). In the mature brain, [Cl]i is maintained at low levels by chloride extrusion, which renders γ-aminobutyric acid (GABA) hyperpolarizing (2) and counteracts activity-dependent chloride loads (3). GABAergic inhibition in the adult is crucial not only for preventing runaway excitation of glutamatergic cells (4) but also for entraining neuronal assemblies into oscillations underlying cognitive processing (5). However, the capacity of chloride extrusion is low during early brain development (6, 7). Additionally, immature neurons are equipped with chloride uptake mechanisms, particularly with the Na+/K+/2Cl cotransporter NKCC1 (812). NKCC1 contributes to the maintenance of high [Cl]i in the developing brain (13), favoring depolarization through GABAA receptor (GABAAR) activation in vivo (14, 15).When GABA acts as a depolarizing neurotransmitter, neural circuits generate burst-like spontaneous activity (1620), which is crucial for their developmental refinement (2124). In vitro evidence indicates that GABAergic interneurons promote neuronal synchrony in an NKCC1-dependent manner (10, 12, 2528). However, the in vivo developmental functions of NKCC1 are far from understood (29, 30). One fundamental question is to what extent NKCC1 and GABAergic depolarization supports correlated spontaneous activity in the neonatal brain. In the neocortex, GABA imposes spatiotemporal inhibition on network activity already in the neonatal period (14, 25, 31, 32). Whether a similar situation applies to other brain regions is unknown, as two recent chemo- and optogenetic studies in the hippocampus yielded opposing results (25, 33). Manipulations of the chloride driving force are potentially suited to resolve these divergent findings, but pharmacological (3436) or conventional knockout (10, 11, 37) strategies suffer from unspecific effects that complicate interpretations.Here, we overcome this limitation by selectively deleting Slc12a2 (encoding NKCC1) from telencephalic glutamatergic neurons. We show that chloride uptake via NKCC1 promotes synchronized activity in acute hippocampal slices, but has weak and event type-dependent effects in CA1 in vivo. Long-term loss of NKCC1 leads to subtle changes of network dynamics in the adult, leaving synaptic development unperturbed and behavioral performance intact. Our data suggest that NKCC1-dependent chloride uptake is largely dispensable for several key aspects of hippocampal development in vivo.  相似文献   

15.
16.
17.
Natural killer (NK) cells are major antileukemic immune effectors. Leukemic blasts have a negative impact on NK cell function and promote the emergence of phenotypically and functionally impaired NK cells. In the current work, we highlight an accumulation of CD56CD16+ unconventional NK cells in acute myeloid leukemia (AML), an aberrant subset initially described as being elevated in patients chronically infected with HIV-1. Deep phenotyping of NK cells was performed using peripheral blood from patients with newly diagnosed AML (n = 48, HEMATOBIO cohort, NCT02320656) and healthy subjects (n = 18) by mass cytometry. We showed evidence of a moderate to drastic accumulation of CD56CD16+ unconventional NK cells in 27% of patients. These NK cells displayed decreased expression of NKG2A as well as the triggering receptors NKp30 and NKp46, in line with previous observations in HIV-infected patients. High-dimensional characterization of these NK cells highlighted a decreased expression of three additional major triggering receptors required for NK cell activation, NKG2D, DNAM-1, and CD96. A high proportion of CD56CD16+ NK cells at diagnosis was associated with an adverse clinical outcome and decreased overall survival (HR = 0.13; P = 0.0002) and event-free survival (HR = 0.33; P = 0.018) and retained statistical significance in multivariate analysis. Pseudotime analysis of the NK cell compartment highlighted a disruption of the maturation process, with a bifurcation from conventional NK cells toward CD56CD16+ NK cells. Overall, our data suggest that the accumulation of CD56CD16+ NK cells may be the consequence of immune escape from innate immunity during AML progression.

Natural killer (NK) cells are critical cytotoxic effectors involved in leukemic blast recognition, tumor cell clearance, and maintenance of long-term remission (1). NK cells directly kill target cells without prior sensitization, enabling lysis of cells stressed by viral infections or tumor transformation. NK cells are divided into different functional subsets according to CD56 and CD16 expression (24). CD56bright NK cells are the most immature NK cells found in peripheral blood. This subset is less cytotoxic than mature NK cells and secretes high amounts of chemokines and cytokines such as IFNγ and TNFα. These cytokines have a major effect on the infected or tumor target cells and play a critical role in orchestration of the adaptive immune response through dendritic cell activation. CD56dimCD16+ NK cells, which account for the majority of circulating human NK cells, are the most cytotoxic NK cells. NK cell activation is finely tuned by integration of signals from inhibitory and triggering receptors, in particular, those of NKp30, NKp46 and NKp44, DNAM-1, and NKG2D (5). Upon target recognition, CD56dimCD16+ NK cells release perforin and granzyme granules and mediate antibody-dependent cellular cytotoxicity through CD16 (FcɣRIII) to clear transformed cells.NK cells are a major component of the antileukemic immune response, and NK cell alterations have been associated with adverse clinical outcomes in acute myeloid leukemia (AML) (69). Therefore, it is crucial to better characterize AML-induced NK cell alterations in order to optimize NK cell–targeted therapies. During AML progression, NK cell functions are deeply altered, with decreased expression of NK cell–triggering receptors and reduced cytotoxic functions as well as impaired NK cell maturation (6, 913). Cancer-induced NK cell impairment occurs through various mechanisms of immune escape, including shedding and release of ligands for NK cell–triggering receptors; release of immunosuppressive soluble factors such as TGFβ, adenosine, PGE2, or L-kynurenine; and interference with NK cell development, among others (14).Interestingly, these mechanisms of immune evasion are also seen to some extent in chronic viral infections, notably HIV (2). In patients with HIV, NK cell functional anergy is mediated by the release of inflammatory cytokines and TGFβ, the presence of MHClow target cells, and the shedding of ligands for NK cell–triggering receptors (2). As a consequence, some phenotypical alterations described in cancer patients are also induced by chronic HIV infections, with decreased expression of major triggering receptors such as NKp30, NKp46, and NKp44 (15, 16); decreased expression of CD16 (17); and increased expression of inhibitory receptors such as T cell immunoreceptor with Ig and ITIM domains (TIGIT) (18) all observed. In addition, patients with HIV display an accumulation of CD56CD16+ unconventional NK cells, a highly dysfunctional NK cell subset (19, 20). Mechanisms leading to the loss of CD56 are still poorly described, and the origin of this subset of CD56 NK cells is still unknown. To date, two hypotheses have been considered: CD56 NK cells could be terminally differentiated cells arising from a mixed population of mature NK cells with altered characteristics or could expand from a pool of immature precursor NK cells (21). Expansion of CD56CD16+ NK cells is mainly observed in viral noncontrollers (19, 20). Indeed, CD56 is an important adhesion molecule involved in NK cell development, motility, and pathogen recognition (2227). CD56 is also required for the formation of the immunological synapse between NK cells and target cells, lytic functions, and cytokine production (26, 28). As a consequence, CD56CD16+ NK cells display lower degranulation capacities and decreased expression of triggering receptors, perforin, and granzyme B, dramatically reducing their cytotoxic potential, notably against tumor target cells (2, 19, 20, 29, 30). In line with this loss of the cytotoxic functions against tumor cells, patients with concomitant Burkitt lymphoma and Epstein-Barr virus infection display a dramatic increase of CD56CD16+ NK cells (30), which could represent an important hallmark of escape to NK cell immunosurveillance in virus-driven hematological malignancies.To our knowledge, this population has not been characterized in the context of nonvirally induced hematological malignancies. In the present work, we investigated the presence of this population of unconventional NK cells in patients with AML, its phenotypical characteristics, and the consequences of its accumulation on disease control. Finally, we explored NK cell developmental trajectories leading to the emergence of this phenotype.  相似文献   

18.
We previously described a new osteogenic growth factor, osteolectin/Clec11a, which is required for the maintenance of skeletal bone mass during adulthood. Osteolectin binds to Integrin α11 (Itga11), promoting Wnt pathway activation and osteogenic differentiation by leptin receptor+ (LepR+) stromal cells in the bone marrow. Parathyroid hormone (PTH) and sclerostin inhibitor (SOSTi) are bone anabolic agents that are administered to patients with osteoporosis. Here we tested whether osteolectin mediates the effects of PTH or SOSTi on bone formation. We discovered that PTH promoted Osteolectin expression by bone marrow stromal cells within hours of administration and that PTH treatment increased serum osteolectin levels in mice and humans. Osteolectin deficiency in mice attenuated Wnt pathway activation by PTH in bone marrow stromal cells and reduced the osteogenic response to PTH in vitro and in vivo. In contrast, SOSTi did not affect serum osteolectin levels and osteolectin was not required for SOSTi-induced bone formation. Combined administration of osteolectin and PTH, but not osteolectin and SOSTi, additively increased bone volume. PTH thus promotes osteolectin expression and osteolectin mediates part of the effect of PTH on bone formation.

The maintenance and repair of the skeleton require the generation of new bone cells throughout adult life. Osteoblasts are relatively short-lived cells that are constantly regenerated, partly by skeletal stem cells within the bone marrow (1). The main source of new osteoblasts in adult bone marrow is leptin receptor-expressing (LepR+) stromal cells (24). These cells include the multipotent skeletal stem cells that give rise to the fibroblast colony-forming cells (CFU-Fs) in the bone marrow (2), as well as restricted osteogenic progenitors (5) and adipocyte progenitors (68). LepR+ cells are a major source of osteoblasts for fracture repair (2) and growth factors for hematopoietic stem cell maintenance (911).One growth factor synthesized by LepR+ cells, as well as osteoblasts and osteocytes, is osteolectin/Clec11a, a secreted glycoprotein of the C-type lectin domain superfamily (5, 12, 13). Osteolectin is an osteogenic factor that promotes the maintenance of the adult skeleton by promoting the differentiation of LepR+ cells into osteoblasts. Osteolectin acts by binding to integrin α11β1, which is selectively expressed by LepR+ cells and osteoblasts, activating the Wnt pathway (12). Deficiency for either Osteolectin or Itga11 (the gene that encodes integrin α11) reduces osteogenesis during adulthood and causes early-onset osteoporosis in mice (12, 13). Recombinant osteolectin promotes osteogenic differentiation by bone marrow stromal cells in culture and daily injection of mice with osteolectin systemically promotes bone formation.Osteoporosis is a progressive condition characterized by reduced bone mass and increased fracture risk (14). Several factors contribute to osteoporosis development, including aging, estrogen insufficiency, mechanical unloading, and prolonged glucocorticoid use (14). Existing therapies include antiresorptive agents that slow bone loss, such as bisphosphonates (15, 16) and estrogens (17), and anabolic agents that increase bone formation, such as parathyroid hormone (PTH) (18), PTH-related protein (19), and sclerostin inhibitor (SOSTi) (20). While these therapies increase bone mass and reduce fracture risk, they are not a cure.PTH promotes both anabolic and catabolic bone remodeling (2124). PTH is synthesized by the parathyroid gland and regulates serum calcium levels, partly by regulating bone formation and bone resorption (2325). PTH1R is a PTH receptor (26, 27) that is strongly expressed by LepR+ bone marrow stromal cells (8, 2830). Recombinant human PTH (Teriparatide; amino acids 1 to 34) and synthetic PTH-related protein (Abaloparatide) are approved by the US Food and Drug Administration (FDA) for the treatment of osteoporosis (19, 31). Daily (intermittent) administration of PTH increases bone mass by promoting the differentiation of osteoblast progenitors, inhibiting osteoblast and osteocyte apoptosis, and reducing sclerostin levels (3235). PTH promotes osteoblast differentiation by activating Wnt and BMP signaling in bone marrow stromal cells (28, 36, 37), although the mechanisms by which it regulates Wnt pathway activation are complex and uncertain (38).Sclerostin is a secreted glycoprotein that inhibits Wnt pathway activation by binding to LRP5/6, a widely expressed Wnt receptor (7, 8), reducing bone formation (39, 40). Sclerostin is secreted by osteocytes (8, 41), negatively regulating bone formation by inhibiting the differentiation of osteoblasts (41, 42). SOSTi (Romosozumab) is a humanized monoclonal antibody that binds sclerostin, preventing binding to LRP5/6 and increasing Wnt pathway activation and bone formation (43). It is FDA-approved for the treatment of osteoporosis (20, 44) and has activity in rodents in addition to humans (45, 46).The discovery that osteolectin is a bone-forming growth factor raises the question of whether it mediates the effects of PTH or SOSTi on osteogenesis.  相似文献   

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