首页 | 官方网站   微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 625 毫秒
1.
2.
Skeletal muscle and bone homeostasis are regulated by members of the myostatin/GDF-11/activin branch of the transforming growth factor-β superfamily, which share many regulatory components, including inhibitory extracellular binding proteins and receptors that mediate signaling. Here, we present the results of genetic studies demonstrating a critical role for the binding protein follistatin (FST) in regulating both skeletal muscle and bone. Using an allelic series corresponding to varying expression levels of endogenous Fst, we show that FST acts in an exquisitely dose-dependent manner to regulate both muscle mass and bone density. Moreover, by employing a genetic strategy to target Fst expression only in the posterior (caudal) region of the animal, we show that the effects of Fst loss are mostly restricted to the posterior region, implying that locally produced FST plays a much more important role than circulating FST with respect to regulation of muscle and bone. Finally, we show that targeting receptors for these ligands specifically in osteoblasts leads to dramatic increases in bone mass, with trabecular bone volume fraction being increased by 12- to 13-fold and bone mineral density being increased by 8- to 9-fold in humeri, femurs, and lumbar vertebrae. These findings demonstrate that bone, like muscle, has an enormous inherent capacity for growth that is normally kept in check by this signaling system and suggest that the extent to which this regulatory mechanism may be used throughout the body to regulate tissue mass may be more significant than previously appreciated.

Myostatin (MSTN) is a transforming growth factor-β (TGF-β) superfamily member that normally acts to limit skeletal muscle mass (1). Mice lacking MSTN exhibit dramatic increases in skeletal muscle mass throughout the body, with individual muscles growing to about twice the normal size. The amino acid sequence of MSTN has been strongly conserved through evolution (2), and engineered or naturally occurring mutations in the MSTN gene have been shown to lead to increased muscling in many other species as well, including cattle (24), sheep (5), dogs (6), rabbits (7), rats (8), swine (9), goats (10), and humans (11). MSTN is regulated by various extracellular binding proteins, including follistatin (FST) (12), FSTL-3 (13), GASP-1 (14), and GASP-2 (15, 16), as well as the MSTN propeptide, which maintains MSTN in an inactive, latent state (12, 1719). MSTN signals initially by binding to the activin type 2 receptors ACVR2 and ACVR2B (12, 2022), followed by engagement of the type 1 receptors ALK4 and ALK5 (22, 23).The function of MSTN as a negative regulator of muscle growth is partially redundant with that of another TGF-β family member, activin A (20, 2427), which shares many regulatory and signaling components with MSTN. Indeed, one of these components, FST, was originally identified for its ability to inhibit secretion of follicle-stimulating hormone (FSH) by cultured pituitary cells (28), and subsequent work showed that FST is capable of binding and inhibiting activins (29), which are capable of signaling to pituitary gonadotrophs to induce FSH secretion (30). FST undergoes alternative splicing to generate two isoforms, the full-length FST315 and a carboxyl-terminal truncated FST288 (31). A third form, FST303, is derived from proteolytic cleavage of the C-terminal domain. All of the FST isoforms contain a heparin-binding domain that mediates binding to cell-surface proteoglycans. The presence of the C-terminal acidic tail in FST315, however, appears to neutralize the basic residues present in the heparin-binding domain and, as a result, FST315 binds poorly to proteoglycans and is the predominant form of FST in the circulation. FST288, which lacks the C-terminal 26–amino acid extension, tends to remain locally sequestered following secretion.Based on the existence of these multiple isoforms and their differential biodistribution following secretion, a major question has been whether the mode of action of FST is primarily local, regulating signaling by target ligands at or near the site of FST synthesis, or whether circulating FST can influence signaling to tissues distant from its site of synthesis. This question is relevant not only in terms of understanding the mechanism of action of FST but also in terms of interpreting human studies seeking associations between circulating FST levels and various physiological and pathological states. Here, we use genetic approaches in mice to address this fundamental question with respect to the roles of FST and this regulatory system in regulating two tissues, skeletal muscle and bone.  相似文献   

3.
4.
The immunosuppressive and hypoxic tumor microenvironment (TME) remains a major obstacle to impede cancer immunotherapy. Here, we showed that elevated levels of Delta-like 1 (DLL1) in the breast and lung TME induced long-term tumor vascular normalization to alleviate tumor hypoxia and promoted the accumulation of interferon γ (IFN-γ)–expressing CD8+ T cells and the polarization of M1-like macrophages. Moreover, increased DLL1 levels in the TME sensitized anti-cytotoxic T lymphocyte–associated protein 4 (anti-CTLA4) treatment in its resistant tumors, resulting in tumor regression and prolonged survival. Mechanically, in vivo depletion of CD8+ T cells or host IFN-γ deficiency reversed tumor growth inhibition and abrogated DLL1-induced tumor vascular normalization without affecting DLL1-mediated macrophage polarization. Together, these results demonstrate that elevated DLL1 levels in the TME promote durable tumor vascular normalization in a CD8+ T cell– and IFN-γ–dependent manner and potentiate anti-CTLA4 therapy. Our findings unveil DLL1 as a potential target to persistently normalize the TME to facilitate cancer immunotherapy.

One of the major challenges currently facing cancer treatments is the aberrant tumor microenvironment (TME), characterized as hypoxia, immunosuppression, acidity, and high interstitial fluid pressure (IFP) (15). These properties render tumors resistant to many kinds of cancer treatment modalities. High IFP prevents the penetration and distribution of drug agents into the tumor parenchyma, while hypoxia compromises the effectiveness of chemotherapy and radiotherapy because both treatment modalities often require reactive oxygen species to evoke antitumor activities (4, 6). In addition, hypoxia induces the secretion of multiple immune inhibitory factors and promotes the accumulation of immune regulatory cell populations, such as transforming growth factor-β (TGF-β), interleukin 10 (IL10), myeloid-derived suppressor cells (MDSCs), M2-like tumor-associated macrophages (M2-TAMs), and regulatory T cells (Tregs) (1, 3, 79). Thus, the hypoxic and immunosuppressive TME hinders cancer immunotherapy to efficiently eradicate cancer cells.Emerging evidence suggests that the abnormal tumor vasculature contributes largely to the aberrant TME (1, 3, 4). Tumor blood vessels are tortuous, dilated, and leaky with low pericyte coverage. The resulting blood flow is often static and fluctuated and therefore creates a hypoxic and acidic TME with high IFP (4). Therefore, tumor vascular normalization has been proposed as a promising approach to alleviate the aberrances within the TME, thus enhancing the efficacy of a range of cancer treatment modalities, including chemotherapy, radiotherapy, and immunotherapy (1018). Vascular endothelial growth factor (VEGF) ligands and receptors constitute one of the most potent proangiogenic signaling pathways (19). Various VEGF signaling inhibitors, such as Bevacizumab and Cediranib, have been approved to treat several types of cancers. VEGF signaling inhibitors can induce tumor vascular normalization; however, the duration of the normalization is usually transient, and therefore, the improvement to the concurrent chemotherapy and immunotherapy is marginal (4, 1921). In addition, many kinds of cancer are intrinsically resistant to VEGF signaling targeted therapy (4, 19). Thus, novel approaches are needed to induce tumor vascular normalization for longer periods and in broad tumor types.The evolutionarily conserved Notch signaling pathway plays critical roles in cell differentiation and blood vessel formation. The Notch signaling pathway consists of four Notch receptors (Notch 1 to 4) and four ligands (Jagged1, Jagged2, Delta-like 1 [DLL1], and DLL4) in murine (22). Both Notch receptors and ligands are membrane proteins. DLL1, DLL4, and Jagged1 have been shown to express in endothelial cells and play important roles in vascular development and postnatal vessel formation (23, 24). DLL1 and DLL4 are also associated with tumor angiogenesis (2426). DLL4 is usually expressed in tumor endothelial cells but rarely in tumor cells (27, 28). Blockade of DLL4 suppresses tumor growth through the induction of nonfunctional tumor vessel formation (24, 25, 29). Thus, activation of DLL4/Notch signaling has the potential to increase tumor vascular maturation. Indeed, higher expression of DLL4 in bladder tumor endothelial cells was correlated with vessel maturation (30). Unfortunately, long-term DLL4 blockade led to vascular neoplasms, and persistent activation of DLL4/Notch signaling promoted T cell acute lymphoblastic leukemia (T-ALL) (3133).Because of these potential safety concerns of chronic blockade or activation of DLL4/Notch signaling, we proposed instead to remodel tumor vessels via the activation of DLL1/Notch signaling. In contrast to the extensive attention of DLL4 in tumor angiogenesis, the roles of DLL1 in tumor vessel formation is largely unknown. Here, we showed that overexpression of DLL1 in EO771 breast and LAP0297 lung tumor cells not only induced durable tumor vascular normalization but also stimulated CD8+ T cell activities. Interestingly, in vivo depletion of CD8+ T cells prior to tumor implantation or host IFN-γ deficiency abrogated the effects of DLL1 overexpression on tumor vessels, suggesting that selective activation of DLL1/Notch signaling induces long-term tumor vascular normalization via T cell activation. Moreover, DLL1/Notch signaling activation in combination with anti-CTLA4 therapy prolonged survival. Thus, this study uncovered DLL1 as a potential target to induce long-term tumor vascular normalization to enhance cancer immunotherapy.  相似文献   

5.
Little is known about the cellular signals that organize synapse formation. To explore what signaling pathways may be involved, we employed heterologous synapse formation assays in which a synaptic adhesion molecule expressed in a nonneuronal cell induces pre- or postsynaptic specializations in cocultured neurons. We found that interfering pharmacologically with microtubules or actin filaments impaired heterologous synapse formation, whereas blocking protein synthesis had no effect. Unexpectedly, pharmacological inhibition of c-jun N-terminal kinases (JNKs), protein kinase-A (PKA), or AKT kinases also suppressed heterologous synapse formation, while inhibition of other tested signaling pathways—such as MAP kinases or protein kinase C—did not alter heterologous synapse formation. JNK and PKA inhibitors suppressed formation of both pre- and postsynaptic specializations, whereas AKT inhibitors impaired formation of post- but not presynaptic specializations. To independently test whether heterologous synapse formation depends on AKT signaling, we targeted PTEN, an enzyme that hydrolyzes phosphatidylinositol 3-phosphate and thereby prevents AKT kinase activation, to postsynaptic sites by fusing PTEN to Homer1. Targeting PTEN to postsynaptic specializations impaired heterologous postsynaptic synapse formation induced by presynaptic adhesion molecules, such as neurexins and additionally decreased excitatory synapse function in cultured neurons. Taken together, our results suggest that heterologous synapse formation is driven via a multifaceted and multistage kinase network, with diverse signals organizing pre- and postsynaptic specializations.

Synapse formation is the universal process that underlies construction of all of the brain’s circuits, but little is known about its mechanisms. Unknown signaling pathways presumably organize synapses, but what pathways are involved remains unclear. Synapse formation likely requires interactions between pre- and postsynaptic neurons via adhesion molecules that transmit bidirectional signals to pre- and postsynaptic neurons and organize pre- and postsynaptic specializations (reviewed in refs. 13). Synapses exhibit canonical features that include a presynaptic side that releases neurotransmitters rapidly and transiently and a postsynaptic side that recognizes these neurotransmitters. Interestingly, only the presynaptic side of a synapse harbors canonical features that are shared by all synapses, such as synaptic vesicles and active zones with the same components in excitatory and inhibitory synapses. In contrast, the postsynaptic sides differ dramatically between excitatory and inhibitory synapses. Even excitatory and inhibitory neurotransmitter receptors exhibit no homology, and few if any molecular components are shared among excitatory and inhibitory postsynaptic specializations.At present, it is unknown what intracellular signaling pathways are involved in the assembly of pre- and postsynaptic specializations, whether different types of signaling pathways exist for pre- vs. postsynaptic specializations, and how excitatory vs. inhibitory synapses are organized. In the present study, we chose the heterologous synapse formation assay as an approach in order to begin to address these fundamental questions (4). In the heterologous synapse formation assay, nonneuronal cells, such as HEK293T cells, express a synaptic adhesion molecule that then induces pre- or postsynaptic specializations when these nonneuronal cells are cocultured with neurons (59). For example, if a postsynaptic adhesion molecule, such as neuroligin-1 (Nlgn1) or latrophilin-3, is expressed in HEK293T cells, and the HEK293T cells are cocultured with neurons, these neurons form presynaptic specializations on the HEK293T cells (5, 10). If, conversely, a presynaptic adhesion molecule, such as a neurexin or teneurin, is expressed in HEK293T cells, postsynaptic specializations are induced in cocultured neurons (8, 9, 11).Many adhesion molecules have been shown to induce heterologous synapse formation, including neurexins, neuroligins, latrophilins, teneurins, SynCAMs, neuronal pentraxin receptors, SALMs, LAR-type PTPRs, and others (5, 6, 815), suggesting that there are common “synapse signaling” pathways and that the heterologous synapse formation assay nonspecifically transduces different adhesion molecules signals into a response that organizes pre- or postsynaptic specializations. Even engagement of neuronal AMPA-type glutamate receptors by the neuronal pentraxin receptor, when expressed in HEK293T cells, causes organization of postsynaptic specializations in the heterologous synapse formation assay, testifying to the broad nature of the signals that mediate heterologous synapse formation (12). Strikingly, any given adhesion molecule triggers only either pre- or postsynaptic specializations, but not both, indicating signaling specificity. Most adhesion molecules—with the exception of teneurin splice variants (11)—induce both excitatory and inhibitory synaptic specializations at the same time. Heterologous synapses resemble real synapses and are functional (6, 7). Overall, these observations suggest that specific signaling pathways regulate synapse formation and that the heterologous synapse formation assay provides a plausible and practical paradigm to dissect such signaling pathways, even though it represents an artificial system that lacks much of the specificity of physiological synapse formation.In the present study, we have employed pharmacological inhibitors and molecular interventions to probe the nature of the signals mediating heterologous synapse formation. Our data reveal that multiple parallel protein kinase signaling pathways are required for heterologous synapse formation. We identified a role for both JNK and PKA signaling in the formation of pre- and postsynaptic specializations and found that the PI3 kinase pathway is specifically required for the formation of post- but not presynaptic specializations. Thus, our data provide initial insight into the signaling mechanisms underlying heterologous synapse formation that may be relevant for synapse formation in general.  相似文献   

6.
7.
8.
9.
10.
Inflammatory pathologies caused by phagocytes lead to numerous debilitating conditions, including chronic pain and blindness due to age-related macular degeneration. Many members of the sialic acid-binding immunoglobulin-like lectin (Siglec) family are immunoinhibitory receptors whose agonism is an attractive approach for antiinflammatory therapy. Here, we show that synthetic lipid-conjugated glycopolypeptides can insert into cell membranes and engage Siglec receptors in cis, leading to inhibitory signaling. Specifically, we construct a cis-binding agonist of Siglec-9 and show that it modulates mitogen-activated protein kinase (MAPK) signaling in reporter cell lines, immortalized macrophage and microglial cell lines, and primary human macrophages. Thus, these cis-binding agonists of Siglecs present a method for therapeutic suppression of immune cell reactivity.

Sialic acid-binding immunoglobulin (IgG)-like lectins (Siglecs) are a family of immune checkpoint receptors that are on all classes of immune cells (15). Siglecs bind various sialoglycan ligands and deliver signals to the immune cells that report on whether the target is healthy or damaged, “self” or “nonself.” Of the 14 human Siglecs, 9 contain cytosolic inhibitory signaling domains. Accordingly, engagement of these inhibitory Siglecs by sialoglycans suppresses the activity of the immune cell, leading to an antiinflammatory effect. In this regard, inhibitory Siglecs have functional parallels with the T cell checkpoint receptors CTLA-4 and PD-1 (69). As with these clinically established targets for cancer immune therapy, there has been a recent surge of interest in antagonizing Siglecs to potentiate immune cell reactivity toward cancer (10). Conversely, engagement of Siglecs with agonist antibodies can suppress immune cell reactivity in the context of antiinflammatory therapy. This approach has been explored to achieve B cell suppression in lupus patients by agonism of CD22 (Siglec-2) (11, 12), and to deplete eosinophils for treatment of eosinophilic gastroenteritis by agonism of Siglec-8 (13). Similarly, a CD24 fusion protein has been investigated clinically as a Siglec-10 agonist for both graft-versus-host disease and viral infection (14, 15).Traditionally, Siglec ligands have been studied as functioning in trans, that is, on an adjacent cell (1618), or as soluble clustering agents (9, 19). In contrast to these mechanisms of action, a growing body of work suggests that cis ligands for Siglecs (i.e., sialoglycans that reside on the same cell membrane) cluster these receptors and maintain a basal level of inhibitory signaling that increases the threshold for immune cell activation. Both Bassik and coworkers (20) and Wyss-Coray and coworkers (21) have linked the depletion of cis Siglec ligands with increased activity of macrophages and microglia, and other studies have shown that a metabolic blockade of sialic acid renders phagocytes more prone to activation (22).Synthetic ligands are a promising class of Siglec agonists (17, 23, 24). Many examples rely on clustering architectures (e.g., sialopolymers, nanoparticles, liposomes) to induce their effect (19, 2326). Indeed, we have previously used glycopolymers to study the effects of Siglec engagement in trans on natural killer (NK) cell activity (16). We and other researchers have employed glycopolymers (16, 23), glycan-remodeling enzymes (27, 28), chemical inhibitors of glycan biosynthesis (22), and mucin overexpression constructs (29, 30) to modulate the cell-surface levels of Siglec ligands. However, current approaches lack specificity for a given Siglec.We hypothesized that Siglec-specific cis-binding sialoglycans displayed on immune cell surfaces could dampen immune cell activity with potential therapeutic applications. Here we test this notion with the synthesis of membrane-tethered cis-binding agonists of Siglec-9 (Fig. 1). Macrophages and microglia widely express Siglec-9 and are responsible for numerous pathologies including age-related inflammation (31), macular degeneration (32), neural inflammation (33), and chronic obstructive pulmonary disease (34). We designed and developed a lipid-linked glycopolypeptide scaffold bearing glycans that are selective Siglec-9 ligands (pS9L-lipid). We show that pS9L-lipid inserts into macrophage membranes, binds Siglec-9 specifically and in cis, and induces Siglec-9 signaling to suppress macrophage activity. By contrast, a lipid-free soluble analog (pS9L-sol) binds Siglec-9 but does not agonize Siglec-9 or modulate macrophage activity. Membrane-tethered glycopolypeptides are thus a potential therapeutic modality for inhibiting phagocyte activity.Open in a separate windowFig. 1.Lipid-tethered glycopolypeptides cluster and agonize Siglecs in cis on effector cells. (A) Immune cells express activating receptors that stimulate inflammatory signaling. (B) Clustering of Siglec-9 by cis-binding agonists stimulates inhibitory signaling that quenches activation.  相似文献   

11.
Osteoarthritis (OA), the leading cause of pain and disability worldwide, disproportionally affects individuals with obesity. The mechanisms by which obesity leads to the onset and progression of OA are unclear due to the complex interactions among the metabolic, biomechanical, and inflammatory factors that accompany increased adiposity. We used a murine preclinical model of lipodystrophy (LD) to examine the direct contribution of adipose tissue to OA. Knee joints of LD mice were protected from spontaneous or posttraumatic OA, on either a chow or high-fat diet, despite similar body weight and the presence of systemic inflammation. These findings indicate that adipose tissue itself plays a critical role in the pathophysiology of OA. Susceptibility to posttraumatic OA was reintroduced into LD mice using implantation of a small adipose tissue depot derived from wild-type animals or mouse embryonic fibroblasts that undergo spontaneous adipogenesis, implicating paracrine signaling from fat, rather than body weight, as a mediator of joint degeneration.

Osteoarthritis (OA) is the leading cause of pain and disability worldwide and is associated with increased all-cause mortality and cardiovascular disease (1, 2). OA is strongly associated with obesity, suggesting that either increased biomechanical joint loading or systemic inflammation and metabolic dysfunction related to obesity are responsible for joint degeneration (1, 2). However, increasing evidence is mounting that changes in biomechanical loading due to increased body mass do not account for the severity of obesity-induced knee OA (19). These observations suggest that other factors related to the presence of adipose tissue and adipose tissue-derived cytokines—termed adipokines—play critical roles in this process and other musculoskeletal conditions (1, 2, 6, 7, 10). As there are presently no disease-modifying OA drugs available, direct evidence linking adipose tissue and cartilage health could provide important mechanistic insight into the natural history of OA and obesity and therefore guide the development and translation of novel OA therapeutic strategies designed to preserve joint health.The exact contribution of the adipokine-signaling network in OA has been difficult to determine due to the complex interactions among metabolic, biomechanical, and inflammatory factors related to obesity (11). To date, the link between increased adipose tissue mass and OA pathogenesis has largely been correlative (6, 7, 12), and, as such, the direct effect of adipose tissue and the adipokines it releases has been difficult to separate from other factors such as dietary composition or excess body mass in the context of obesity, which is most commonly caused by excessive nutrition (2, 6, 7). In particular, leptin, a proinflammatory adipokine and satiety hormone secreted proportionally to adipose tissue mass is most consistently increased in obesity-induced OA (1), and leptin knockout mice are protected from OA (6, 7). However, it remains to be determined whether leptin directly contributes to OA pathogenesis, independent of its effect on metabolism (and weight). Additional adipokines that have been implicated in the onset and progression of OA include adiponectin, resistin, visfatin, chimerin, and inflammatory cytokines such as interleukin-1 (IL-1), IL-6, and tumor necrosis factor-α (TNF-α) (13). The infrapatellar fat pad represents a local source of adipokines within the knee joint, but several studies indicate strong correlations with visceral adipose tissue, outside of the joint organ system, with OA severity (14). Furthermore, adipokine receptors are found on almost all cells within the joint and, therefore, could directly contribute to OA pathogenesis through synovitis, cartilage damage, and bone remodeling (13). The role of other adipokines (15) in OA pathogenesis remains to be determined, as it has been difficult to separate and directly test the role of adipokines from other biomechanical, inflammatory, and metabolic factors that contribute to OA pathogenesis.To directly investigate the mechanisms by which adipose tissue affects OA, we used a transgenic mouse with lipodystrophy (LD) that completely lacks adipose tissue and, therefore, adipokine signaling. The LD model system affords the unique opportunity to directly examine the effects of adipose tissue and its secretory factors on musculoskeletal pathology without the confounding effect of diet (16, 17). While LD mice completely lack adipose tissue depots, they demonstrate similar body mass to wild-type (WT) controls on a chow diet (12, 1619). These characteristics provide a unique model that can be used to eliminate the factor of loading due to body mass on joint damage and, thus, to directly test the effects of fat and factors secreted by fat on musculoskeletal tissues. Of particular interest, LD mice also exhibit several characteristics that have been associated with OA, including sclerotic bone (11, 20), metabolic derangement (3, 5, 79, 21, 22), and muscle weakness (2). Despite these OA-predisposing features, LD mice are protected from OA and implantation of adipose tissue back into LD mice restores susceptibility to OA—demonstrating a direct relationship between adipose tissue and cartilage health, independent of the effect of obesity on mechanical joint loading.  相似文献   

12.
13.
A transplanted stem cell’s engagement with a pathologic niche is the first step in its restoring homeostasis to that site. Inflammatory chemokines are constitutively produced in such a niche; their binding to receptors on the stem cell helps direct that cell’s “pathotropism.” Neural stem cells (NSCs), which express CXCR4, migrate to sites of CNS injury or degeneration in part because astrocytes and vasculature produce the inflammatory chemokine CXCL12. Binding of CXCL12 to CXCR4 (a G protein-coupled receptor, GPCR) triggers repair processes within the NSC. Although a tool directing NSCs to where needed has been long-sought, one would not inject this chemokine in vivo because undesirable inflammation also follows CXCL12–CXCR4 coupling. Alternatively, we chemically “mutated” CXCL12, creating a CXCR4 agonist that contained a strong pure binding motif linked to a signaling motif devoid of sequences responsible for synthetic functions. This synthetic dual-moity CXCR4 agonist not only elicited more extensive and persistent human NSC migration and distribution than did native CXCL 12, but induced no host inflammation (or other adverse effects); rather, there was predominantly reparative gene expression. When co-administered with transplanted human induced pluripotent stem cell-derived hNSCs in a mouse model of a prototypical neurodegenerative disease, the agonist enhanced migration, dissemination, and integration of donor-derived cells into the diseased cerebral cortex (including as electrophysiologically-active cortical neurons) where their secreted cross-corrective enzyme mediated a therapeutic impact unachieved by cells alone. Such a “designer” cytokine receptor-agonist peptide illustrates that treatments can be controlled and optimized by exploiting fundamental stem cell properties (e.g., “inflammo-attraction”).

A transplanted stem cell’s engagement with a pathologic niche is the first step in cell-mediated restoration of homeostasis to that region, whether by cell replacement, protection, gene delivery, milieu alteration, toxin neutralization, or remodeling (14). Not surprisingly, the more host terrain covered by the stem cells, the greater their impact. We and others found that a propensity for neural stem cells (NSCs) to home in vivo to acutely injured or actively degenerating central nervous system (CNS) regions—a property called “pathotropism” (112), now viewed as central to stem cell biology—is undergirded, at least in part, by the presence of chemokine receptors on the NSC surface, enabling them to follow concentration gradients of inflammatory cytokines constitutively elaborated by pathogenic processes and expressed by reactive astrocytes and injured vascular endothelium within the pathologic niche (59). This engagement of NSC receptors was first described for the prototypical chemokine receptor CXCR4 (C-X-C chemokine receptor type 4; also known as fusin or cluster of differentiation-184 [CD184]) and its unique natural cognate agonist ligand, the inflammatory chemokine CXCL12 (C-X-C motif chemokine ligand-12; also known as stromal cell-derived factor 1α [SDF-1α]) (5), but has since been described for many chemokine receptor-agonist pairings (69). Chemokine receptors belong to a superfamily that is characterized by seven transmembrane GDP-binding protein-coupled receptors (GPCRs) (1321). In addition to their role in mediating inflammatory reactions and immune responses (22, 23), these receptors and their agonists are components of the regulatory axes for hematopoiesis and organogenesis in other systems (21, 24). Therefore, it is not surprising that binding of CXCL12 to CXCR4 mediates not only an inflammatory response, but also triggers within the NSC a series of intracellular processes associated with migration (as well as proliferation, differentiation, survival, and, during early brain development, proper neuronal lamination) (10).A tool directing therapeutic NSCs to where they are needed has long been sought in regenerative medicine (11, 12). While it was appealing to contemplate electively directing reparative NSCs to any desired area by emulating this chemoattractive property through the targeted injection of exogenous recombinant inflammatory cytokines, it ultimately seemed inadvisable to risk increasing toxicity in brains already characterized by excessive and usually inimical inflammation from neurotraumatic or neurodegenerative processes. However, the notion of engaging the homing function of these NSC-borne receptors without triggering that receptor’s undesirable downstream inflammatory signaling [particularly given that the NSCs themselves can exert a therapeutic antiinflammatory action in the diseased region (1, 2)] seemed a promising heretofore unexplored “workaround.”There had already been an impetus to examine the structure–function relationships of CXCR4, known to be the entry route into cells for HIV-1, in order to create CXCR4 antagonists that block viral infection (2530). Antagonists of CXCR4 were also devised to forestall hematopoietic stem cells from homing to the bone marrow, hence prolonging their presence in the peripheral blood (31) to treat blood dyscrasias. An agonist, however, particularly one with discrete and selective actions, had not been contemplated. In other words, if CXCL12 could be stripped of its undesirable actions while preserving its tropic activity, an ideal chemoattractant would be derived.Based on the concept that CXCR4’s functions are conveyed by two distinct molecular “pockets”—one mediating binding (i.e., allowing a ligand to engage CXCR4) and the other mediating signaling (i.e., enabling a ligand, after binding, to trigger CXCR4-mediated intracellular cascades that promote not only inflammation but also migration) (1318)—we performed chemical mutagenesis that should optimize binding while narrowing the spectrum of signaling. We created a simplified de novo peptide agonist of CXCR4 that contained a strong pure binding motif derived from CXCR4’s strongest ligand, viral macrophage inflammatory protein-II (vMIP-II) and linked it to a truncated signaling motif (only 8 amino acid residues) derived from the N terminus of native CXCL12 (19, 20). This synthetic dual-moiety CXCR4 agonist, which is devoid of a large portion of CXCL12’s native sequence (presumably responsible for undesired functions such as inflammation) not only elicited (with great specificity) more extensive and long-lasting human NSC (hNSC) migration and distribution than native CXCL12 (overcoming migratory barriers), but induced no host inflammation (or other adverse effects). Furthermore, because all of the amino acids in the binding motif were in a D-chirality, rendering the peptide resistant to enzymatic degradation, persistence of this benign synthetic agonist in vivo was prolonged. The hNSC’s gene ontology expression profile was predominantly reparative in contrast to inflammatory as promoted by native CXCL12. When coadministered with transplanted human induced pluripotent stem cell (hiPSC)-derived hNSCs (hiPSC derivatives are now known to have muted migration) in a mouse model of a prototypical neurodegenerative disease [the lethal neuropathic lysosomal storage disorder (LSD) Sandhoff disease (29), where hiPSC-hNSC migration is particularly limited], the synthetic agonist enhanced migration, dissemination, and integration of donor-derived cells into the diseased cortex (including as electrophysiologically active cortical neurons), where their secreted cross-corrective enzyme could mediate a histological and functional therapeutic impact in a manner unachieved by transplanting hiPSC-derived cells alone.In introducing such a “designer” cytokine receptor agonist, we hope to offer proof-of-concept that stem cell-mediated treatments can be controlled and optimized by exploiting fundamental stem cell properties (e.g., “inflammo-attraction”) to alter a niche and augment specific actions. Additionally, when agonists are strategically designed, the various functions of chemokine receptors (and likely other GCPRs) may be divorced. We demonstrate that such a strategy might be used safely and effectively to direct cells to needed regions and broaden their chimerism. We discuss the future implications and uses within the life sciences of such a chemical engineering approach.  相似文献   

14.
Various neuromodulation approaches have been employed to alter neuronal spiking activity and thus regulate brain functions and alleviate neurological disorders. Infrared neural stimulation (INS) could be a potential approach for neuromodulation because it requires no tissue contact and possesses a high spatial resolution. However, the risk of overheating and an unclear mechanism hamper its application. Here we show that midinfrared stimulation (MIRS) with a specific wavelength exerts nonthermal, long-distance, and reversible modulatory effects on ion channel activity, neuronal signaling, and sensorimotor behavior. Patch-clamp recording from mouse neocortical pyramidal cells revealed that MIRS readily provides gain control over spiking activities, inhibiting spiking responses to weak inputs but enhancing those to strong inputs. MIRS also shortens action potential (AP) waveforms by accelerating its repolarization, through an increase in voltage-gated K+ (but not Na+) currents. Molecular dynamics simulations further revealed that MIRS-induced resonance vibration of –C=O bonds at the K+ channel ion selectivity filter contributes to the K+ current increase. Importantly, these effects are readily reversible and independent of temperature increase. At the behavioral level in larval zebrafish, MIRS modulates startle responses by sharply increasing the slope of the sensorimotor input–output curve. Therefore, MIRS represents a promising neuromodulation approach suitable for clinical application.

Many forms of neuromodulation have been used for the regulation of brain functions and the treatment of brain disorders. Some physical approaches, such as electrical, magnetic, and optical (electromagnetic; EM) stimulation could be employed to manipulate neural spiking activity and achieve neuromodulation. Among them, deep-brain electrical stimulation has become a gold standard treatment for advanced Parkinson’s disease; transcranial magnetic stimulation also generates electrical current in selected brain regions and has been used for mood regulation. In contrast, optical neural stimulation has not been used clinically, largely due to the risk of tissue damage by overheating and unclear mechanisms. Although optogenetic manipulation and stimulation avoid these problems and show cell specificity, the requirement of expression of exogenous genes hinders its use in humans (1).Optical infrared neural stimulation (INS) is emerging as an area of interest for neuromodulation and potential clinical application. INS utilizes brief light pulses to activate excitable cells or tissues in the illumination spot. Previous studies showed that INS could activate peripheral nerves (2), peripheral sensory systems (3, 4), and cardiac tissue (5). In the central nervous system (CNS), initial studies found that INS could evoke neural responses in rat thalamocortical slices in vitro (6) and regulate spiking activity in rodent somatosensory cortex (7) and nonhuman primate visual cortex in vivo (8). Because of its high spatial precision, focal INS has been recently applied to map brain connectomes (9). The underlying mechanism of INS, however, remains poorly understood. The predominant view is the transduction of EM energy to thermal heat (10, 11) will excite the cell, possibly due to heat-induced transmembrane capacitive charge (12), changes in ion channel activity (13), or cell damage (14). Since previous studies tended to choose infrared wavelengths with high water absorption for efficient heat generation, it remains unclear whether INS exerts nonthermal effects on ion channel and neuronal spiking activity.While most studies on infrared stimulation have been conducted at near-infrared wavelengths, whether midinfrared wavelengths can regulate neural function is unknown. Because the frequency of midinfrared light falls into the frequency range of chemical bond vibration (1517), nonlinear resonances may occur within biomolecules (1820), leading to dramatic changes in their conformation and function (21) and thus producing nonthermal effects on biological systems. Ion channel proteins distributed on cell membranes could be potential molecular targets for midinfrared light. Among them, voltage-gated Na+ and K+ channels play critical roles in regulating the initiation and propagation of the action potential (AP), an all-or-none digital signal of neurons (22, 23). They also control the voltage waveform of the AP and thus the size of the postsynaptic response, ensuring analog-mode communication between neurons (2427). It is of interest to know whether midinfrared stimulation (MIRS) can cause conformational change in these channel proteins and consequently regulate neuronal signaling. Previous studies revealed a low absorption of light by water in the midinfrared region from 3.5 to 5.7 μm (28), which could be a potential wavelength range for neuromodulation. Therefore, in this study, we explored whether MIRS with a specific wavelength in this range could exert nonthermal modulatory effects on channel activity, neuronal signaling, and behavior.  相似文献   

15.
Learning and memory are assumed to be supported by mechanisms that involve cholinergic transmission and hippocampal theta. Using G protein–coupled receptor-activation–based acetylcholine sensor (GRABACh3.0) with a fiber-photometric fluorescence readout in mice, we found that cholinergic signaling in the hippocampus increased in parallel with theta/gamma power during walking and REM sleep, while ACh3.0 signal reached a minimum during hippocampal sharp-wave ripples (SPW-R). Unexpectedly, memory performance was impaired in a hippocampus-dependent spontaneous alternation task by selective optogenetic stimulation of medial septal cholinergic neurons when the stimulation was applied in the delay area but not in the central (choice) arm of the maze. Parallel with the decreased performance, optogenetic stimulation decreased the incidence of SPW-Rs. These findings suggest that septo–hippocampal interactions play a task-phase–dependent dual role in the maintenance of memory performance, including not only theta mechanisms but also SPW-Rs.

The neurotransmitter acetylcholine is thought to be critical for hippocampus-dependent declarative memories (1, 2). Reduction in cholinergic neurotransmission, either in Alzheimer’s disease or in experiments with cholinergic antagonists, such as scopolamine, impairs memory function (38). Acetylcholine may bring about its beneficial effects on memory encoding by enhancing theta rhythm oscillations, decreasing recurrent excitation, and increasing synaptic plasticity (911). Conversely, drugs which activate cholinergic receptors enhance learning and, therefore, are a neuropharmacological target for the treatment of memory deficits in Alzheimer’s disease (5, 12, 13).The contribution of cholinergic mechanisms in the acquisition of long-term memories and the role of the hippocampal–entorhinal–cortical interactions are well supported by experimental data (5, 12, 13). In addition, working memory or “short-term” memory is also supported by the hippocampal–entorhinal–prefrontal cortex (1416). Working memory in humans is postulated to be a conscious process to “keep things in mind” transiently (16). In rodents, matching to sample task, spontaneous alternation between reward locations, and the radial maze task have been suggested to function as a homolog of working memory [“working memory like” (17)].Cholinergic activity is a critical requirement for working memory (18, 19) and for sustaining theta oscillations (10, 2022). In support of this contention, theta–gamma coupling and gamma power are significantly higher in the choice arm of the maze, compared with those in the side arms where working memory is no longer needed for correct performance (2326). It has long been hypothesized that working memory is maintained by persistent firing of neurons, which keep the presented items in a transient store in the prefrontal cortex and hippocampal–entorhinal system (2731), although the exact mechanisms are debated (3237). An alternative hypothesis holds that items of working memory are stored in theta-nested gamma cycles (38). Common in these models of working memory is the need for an active, cholinergic system–dependent mechanism (3941). However, in spontaneous alternation tasks, the animals are not moving continuously during the delay, and theta oscillations are not sustained either. During the immobility epochs, theta is replaced by intermittent sharp-wave ripples (SPW-R), yet memory performance does not deteriorate. On the contrary, artificial blockade of SPW-Rs can impair memory performance (42, 43), and prolongation of SPW-Rs improves performance (44). Under the cholinergic hypothesis of working memory, such a result is unexpected.To address the relationship between cholinergic/theta versus SPW-R mechanism in spontaneous alternation, we used a G protein–coupled receptor-activation–based acetylcholine sensor (GRABACh3.0) (45) to monitor acetylcholine (ACh) activity during memory performance in mice. In addition, we optogenetically enhanced cholinergic tone, which suppresses SPW-Rs by a different mechanism than electrically or optogenetically induced silencing of neurons in the hippocampus (43, 44). We show that cholinergic signaling in the hippocampus increases in parallel with theta power/score during walking and rapid eye movement (REM) sleep and reaches a transient minimum during SPW-Rs. Selective optogenetic stimulation of medial septal cholinergic neurons decreased the incidence of SPW-Rs during non-REM sleep (4648), as well as during the delay epoch of a working memory task and impaired memory performance. These findings demonstrate that memory performance is supported by complementary theta and SPW-R mechanisms.  相似文献   

16.
Achieving behavioral goals requires integration of sensory and cognitive information across cortical laminae and cortical regions. How this computation is performed remains unknown. Using local field potential recordings and spectrally resolved conditional Granger causality (cGC) analysis, we mapped visual information flow, and its attentional modulation, between cortical layers within and between macaque brain areas V1 and V4. Stimulus-induced interlaminar information flow within V1 dominated upwardly, channeling information toward supragranular corticocortical output layers. Within V4, information flow dominated from granular to supragranular layers, but interactions between supragranular and infragranular layers dominated downwardly. Low-frequency across-area communication was stronger from V4 to V1, with little layer specificity. Gamma-band communication was stronger in the feedforward V1-to-V4 direction. Attention to the receptive field of V1 decreased communication between all V1 layers, except for granular-to-supragranular layer interactions. Communication within V4, and from V1 to V4, increased with attention across all frequencies. While communication from V4 to V1 was stronger in lower-frequency bands (4 to 25 Hz), attention modulated cGCs from V4 to V1 across all investigated frequencies. Our data show that top-down cognitive processes result in reduced communication within cortical areas, increased feedforward communication across all frequency bands, and increased gamma-band feedback communication.

Goal-directed behavior requires the brain to integrate sensory information with cognitive variables. In neocortical areas, sensory information is conveyed by feedforward connections, while feedback connections convey information about cognitive states and goals. Feedforward and feedback connections rely on separate anatomical pathways and have been proposed to map onto distinct frequency bands of neural population activity (17). It is, however, unknown whether these signals differ across laminae, or how they are communicated between laminae within and between cortical areas.Feedforward connections predominantly terminate in layer IV of sensory cortical areas. This information is passed on to layers II/III and further to layers V/VI, where recurrent inputs to layer II/III arise (811). Cognitive variables affect sensory processing through feedback connections, which predominantly terminate in layer I and V (12), but this termination pattern varies depending on hierarchical distances between areas (13). Feedforward and feedback signals have been proposed to show separate local field potential (LFP) spectral signatures. Feedforward signals have been associated with low-frequency theta- (1, 7) and gamma-band activity, originating and dominating in supragranular layers (17). Feedback signals have been associated with lower-frequency (alpha, beta) band activity, prominent in infragranular layers across the cortical hierarchy (14, 7, 14), although attention-related feedback signals in the gamma-frequency band between frontal eye field (FEF) and V4 have been reported (15). Alpha-related feedback has been linked to active inhibition (16, 17), suggesting that feedback signals, induced by attention to receptive field (RF) locations, should result in reduced alpha power. This occurs in infragranular layers in visual areas (18), but can also be less layer-specific (2). It is thus questionable whether feedback information is transmitted by alpha frequencies because attention, employing feedback, shunts alpha oscillations. In extrastriate sensory areas, attention increases LFP power in the gamma-frequency band (14, 15, 1921), while, in primary visual cortex, attention can increase or decrease LFP power in the gamma-frequency band (1, 14, 21). Many of the above results were obtained by methods which do not provide insight into how these signals differ between laminae within an area, or between laminae across different areas. Thus, it remains unclear whether layer differences in these signals between cortical areas exist, and whether they are differently affected by cognitive goals.To understand how information within and between areas is conveyed as a function of cognitive task, we performed simultaneous laminar recordings in areas V1 and V4 using 16-contact laminar probes while macaque monkeys performed a feature-based spatial attention task (22). We quantified communication between laminae and areas by measuring Granger causality (GC) using locally referenced LFP signals.  相似文献   

17.
18.
Active matter comprises individually driven units that convert locally stored energy into mechanical motion. Interactions between driven units lead to a variety of nonequilibrium collective phenomena in active matter. One of such phenomena is anomalously large density fluctuations, which have been observed in both experiments and theories. Here we show that, on the contrary, density fluctuations in active matter can also be greatly suppressed. Our experiments are carried out with marine algae (Effreniumvoratum), which swim in circles at the air–liquid interfaces with two different eukaryotic flagella. Cell swimming generates fluid flow that leads to effective repulsions between cells in the far field. The long-range nature of such repulsive interactions suppresses density fluctuations and generates disordered hyperuniform states under a wide range of density conditions. Emergence of hyperuniformity and associated scaling exponent are quantitatively reproduced in a numerical model whose main ingredients are effective hydrodynamic interactions and uncorrelated random cell motion. Our results demonstrate the existence of disordered hyperuniform states in active matter and suggest the possibility of using hydrodynamic flow for self-assembly in active matter.

Active matter exists over a wide range of spatial and temporal scales (16) from animal groups (7, 8) to robot swarms (911), to cell colonies and tissues (1216), to cytoskeletal extracts (1720), to man-made microswimmers (2125). Constituent particles in active matter systems are driven out of thermal equilibrium at the individual level; they interact to develop a wealth of intriguing collective phenomena, including clustering (13, 22, 24), flocking (11, 26), swarming (12, 13), spontaneous flow (14, 20), and giant density fluctuations (10, 11). Many of these observed phenomena have been successfully described by particle-based or continuum models (16), which highlight the important roles of both individual motility and interparticle interactions in determining system dynamics.Current active matter research focuses primarily on linearly swimming particles which have a symmetric body and self-propel along one of the symmetry axes. However, a perfect alignment between the propulsion direction and body axis is rarely found in reality. Deviation from such a perfect alignment leads to a persistent curvature in the microswimmer trajectories; examples of such circle microswimmers include anisotropic artificial micromotors (27, 28), self-propelled nematic droplets (29, 30), magnetotactic bacteria and Janus particles in rotating external fields (31, 32), Janus particle in viscoelastic medium (33), and sperm and bacteria near interfaces (34, 35). Chiral motility of circle microswimmers, as predicted by theoretical and numerical investigations, can lead to a range of interesting collective phenomena in circular microswimmers, including vortex structures (36, 37), localization in traps (38), enhanced flocking (39), and hyperuniform states (40). However, experimental verifications of these predictions are limited (32, 35), a situation mainly due to the scarcity of suitable experimental systems.Here we address this challenge by investigating marine algae Effrenium voratum (41, 42). At air–liquid interfaces, E.voratum cells swim in circles via two eukaryotic flagella: a transverse flagellum encircling the cellular anteroposterior axis and a longitudinal one running posteriorly. Over a wide range of densities, circling E.voratum cells self-organize into disordered hyperuniform states with suppressed density fluctuations at large length scales. Hyperuniformity (43, 44) has been considered as a new form of material order which leads to novel functionalities (4549); it has been observed in many systems, including avian photoreceptor patterns (50), amorphous ices (51), amorphous silica (52), ultracold atoms (53), soft matter systems (5461), and stochastic models (6264). Our work demonstrates the existence of hyperuniformity in active matter and shows that hydrodynamic interactions can be used to construct hyperuniform states.  相似文献   

19.
Ciliary neurotrophic factor (CNTF) is a leading therapeutic candidate for several ocular diseases and induces optic nerve regeneration in animal models. Paradoxically, however, although CNTF gene therapy promotes extensive regeneration, recombinant CNTF (rCNTF) has little effect. Because intraocular viral vectors induce inflammation, and because CNTF is an immune modulator, we investigated whether CNTF gene therapy acts indirectly through other immune mediators. The beneficial effects of CNTF gene therapy remained unchanged after deleting CNTF receptor alpha (CNTFRα) in retinal ganglion cells (RGCs), the projection neurons of the retina, but were diminished by depleting neutrophils or by genetically suppressing monocyte infiltration. CNTF gene therapy increased expression of C-C motif chemokine ligand 5 (CCL5) in immune cells and retinal glia, and recombinant CCL5 induced extensive axon regeneration. Conversely, CRISPR-mediated knockdown of the cognate receptor (CCR5) in RGCs or treating wild-type mice with a CCR5 antagonist repressed the effects of CNTF gene therapy. Thus, CCL5 is a previously unrecognized, potent activator of optic nerve regeneration and mediates many of the effects of CNTF gene therapy.

Like most pathways in the mature central nervous system (CNS), the optic nerve cannot regenerate once damaged due in part to cell-extrinsic suppressors of axon growth (1, 2) and the low intrinsic growth capacity of adult retinal ganglion cells (RGCs), the projection neurons of the eye (35). Consequently, traumatic or ischemic optic nerve injury or degenerative diseases such as glaucoma lead to irreversible visual losses. Experimentally, some degree of regeneration can be induced by intraocular inflammation or growth factors expressed by inflammatory cells (610), altering the cell-intrinsic growth potential of RGCs (35), enhancing physiological activity (11, 12), chelating free zinc (13, 14), and other manipulations (1519). However, the extent of regeneration achieved to date remains modest, underlining the need for more effective therapies.Ciliary neurotrophic factor (CNTF) is a leading therapeutic candidate for glaucoma and other ocular diseases (2023). Activation of the downstream signal transduction cascade requires CNTF to bind to CNTF receptor-α (CNTFRα) (24), which leads to recruitment of glycoprotein 130 (gp130) and leukemia inhibitory factor receptor-β (LIFRβ) to form a tripartite receptor complex (25). CNTFRα anchors to the plasma membrane through a glycosylphosphatidylinositol linkage (26) and can be released and become soluble through phospholipase C-mediated cleavage (27). CNTF has been reported to activate STAT3 phosphorylation in retinal neurons, including RGCs, and to promote survival, but it is unknown whether these effects are mediated by direct action of CNTF on RGCs via CNTFRα (28). Our previous studies showed that CNTF promotes axon outgrowth from neonate RGCs in culture (29) but fails to do so in cultured mature RGCs (8) or in vivo (6). Although some studies report that recombinant CNTF (rCNTF) can promote optic nerve regeneration (20, 30, 31), others find little or no effect unless SOCS3 (suppressor of cytokine signaling-3), an inhibitor of the Jak-STAT pathway, is deleted in RGCs (5, 6, 32). In contrast, multiple studies show that adeno-associated virus (AAV)-mediated expression of CNTF in RGCs induces strong regeneration (3340). The basis for the discrepant effects of rCNTF and CNTF gene therapy is unknown but is of considerable interest in view of the many promising clinical and preclinical outcomes obtained with CNTF to date.Because intravitreal virus injections induce inflammation (41), we investigated the possibility that CNTF, a known immune modulator (4244), might act by elevating expression of other immune-derived factors. We report here that the beneficial effects of CNTF gene therapy in fact require immune system activation and elevation of C-C motif chemokine ligand 5 (CCL5). Depletion of neutrophils, global knockout (KO) or RGC-selective deletion of the CCL5 receptor CCR5, or a CCR5 antagonist all suppress the effects of CNTF gene therapy, whereas recombinant CCL5 (rCCL5) promotes axon regeneration and increases RGC survival. These studies point to CCL5 as a potent monotherapy for optic nerve regeneration and to the possibility that other applications of CNTF and other forms of gene therapy might similarly act indirectly through other factors.  相似文献   

20.
Efficient and faithful replication of the genome is essential to maintain genome stability. Replication is carried out by a multiprotein complex called the replisome, which encounters numerous obstacles to its progression. Failure to bypass these obstacles results in genome instability and may facilitate errors leading to disease. Cells use accessory helicases that help the replisome bypass difficult barriers. All eukaryotes contain the accessory helicase Pif1, which tracks in a 5′–3′ direction on single-stranded DNA and plays a role in genome maintenance processes. Here, we reveal a previously unknown role for Pif1 in replication barrier bypass. We use an in vitro reconstituted Saccharomyces cerevisiae replisome to demonstrate that Pif1 enables the replisome to bypass an inactive (i.e., dead) Cas9 (dCas9) R-loop barrier. Interestingly, dCas9 R-loops targeted to either strand are bypassed with similar efficiency. Furthermore, we employed a single-molecule fluorescence visualization technique to show that Pif1 facilitates this bypass by enabling the simultaneous removal of the dCas9 protein and the R-loop. We propose that Pif1 is a general displacement helicase for replication bypass of both R-loops and protein blocks.

Efficient and faithful replication of the genome is essential to maintain genome stability and is carried out by a multiprotein complex called the replisome (14). There are numerous obstacles to progression of the replisome during the process of chromosome duplication. These obstacles include RNA-DNA hybrids (R-loops), DNA secondary structures, transcribing RNA polymerases, and other tightly bound proteins (59). Failure to bypass these barriers may result in genome instability, which can lead to cellular abnormalities and genetic disease. Cells contain various accessory helicases that help the replisome bypass these difficult barriers (1020). A subset of these helicases act on the opposite strand of the replicative helicase (1, 2, 14, 19).All eukaryotes contain an accessory helicase, Pif1, which tracks in a 5′–3′ direction on single-stranded DNA (ssDNA) (1116). Pif1 is important in pathways such as Okazaki-fragment processing and break-induced repair that require the removal of DNA-binding proteins as well as potential displacement of R-loops (1113, 21, 1518, 2225). Genetic studies and immunoprecipitation pull-down assays indicate that Pif1 interacts with PCNA (the DNA sliding clamp), Pol ε (the leading-strand polymerase), the MCMs (the motor subunits of the replicative helicase CMG), and RPA (the single-stranded DNA-binding protein) (15, 26, 27). Pif1 activity in break-induced repair strongly depends on its interaction with PCNA (26). These interactions with replisomal components suggest that Pif1 could interact with the replisome during replication. In Escherichia coli, the replicative helicase is the DnaB homohexamer that encircles the lagging strand and moves in a 5′–3′ direction (20). E. coli accessory helicases include the monomeric UvrD (helicase II) and Rep, which move in the 3′–5′ direction and operate on the opposite strand from the DnaB hexamer. It is known that these monomeric helicases promote the bypass of barriers during replication such as stalled RNA polymerases (5). The eukaryotic replicative helicase is the 11-subunit CMG (Cdc45, Mcm2–7, GINS) and tracks in the 3′–5′ direction, opposite to the direction of Pif1 (25, 28). Once activated by Mcm10, the MCM motor domains of CMG encircle the leading strand (2932). We hypothesized that, similar to UvrD and Rep in E. coli, Pif1 interacts with the replisome tracking in the opposite direction to enable bypass of replication obstacles.In this report, we use an in vitro reconstituted Saccharomyces cerevisiae replisome to study the role of Pif1 in bypass of a “dead” Cas9 (dCas9), which is a Cas9 protein that is deactivated in DNA cleavage but otherwise fully functional in DNA binding. As with Cas9, dCas9 is a single-turnover enzyme that can be programmed with a guide RNA (gRNA) to target either strand. The dCas9–gRNA complex forms a roadblock consisting of an R-loop and a tightly bound protein (dCas9), a construct that is similar to a stalled RNA polymerase. This roadblock (hereafter dCas9 R-loop) arrests replisomes independent of whether the dCas9 R-loop is targeted to the leading or lagging strand (30). Besides its utility due to its programmable nature (33), the use of the dCas9 R-loop allows us to answer several mechanistic questions. For example, the ability to program the dCas9 R-loop block to any specific sequence enables us to observe whether block removal is different depending on whether the block is on the leading or lagging strand. Furthermore, the inner diameter of CMG can accommodate double-stranded DNA (dsDNA) and possibly an R-loop, but not a dCas9 protein. Using the dCas9 R-loop block allows us to determine the fate of each of its components.Here, we report that Pif1 enables the bypass of the dCas9 R-loop by the replisome. Interestingly, dCas9 R-loops targeted to either the leading or lagging strand are bypassed with similar efficiency. In addition, the PCNA clamp is not required for bypass of the block, indicating that Pif1 does not need to interact with PCNA during bypass of the block. We used a single-molecule fluorescence imaging to show that both the dCas9 and the R-loop are displaced as an intact nucleoprotein complex. We propose that Pif1 is a general displacement helicase for replication bypass of both R-loops and protein blocks.  相似文献   

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-23

京公网安备 11010802026262号