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 593 毫秒
1.
The nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 12 (NLRP12) plays a protective role in intestinal inflammation and carcinogenesis, but the physiological function of this NLR during microbial infection is largely unexplored. Salmonella enterica serovar Typhimurium (S. typhimurium) is a leading cause of food poisoning worldwide. Here, we show that NLRP12-deficient mice were highly resistant to S. typhimurium infection. Salmonella-infected macrophages induced NLRP12-dependent inhibition of NF-κB and ERK activation by suppressing phosphorylation of IκBα and ERK. NLRP12-mediated down-regulation of proinflammatory and antimicrobial molecules prevented efficient clearance of bacterial burden, highlighting a role for NLRP12 as a negative regulator of innate immune signaling during salmonellosis. These results underscore a signaling pathway defined by NLRP12-mediated dampening of host immune defenses that could be exploited by S. typhimurium to persist and survive in the host.The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family consists of a large number of intracellular pathogen recognition receptors that function as sensors of microbial-derived and danger-associated molecules in the cytoplasm of host cells. A subset of NLR proteins, including NLRP1, NLRP3, and NLRC4, activate caspase-1 via the formation of a cytosolic multiprotein complex termed the inflammasome (1). These inflammasome-forming NLRs mediate processing of the proinflammatory cytokines pro–IL-1β and pro–IL-18, which are then secreted by the cell. The non–inflammasome-forming members of the NLR family contribute to regulation of other key inflammatory pathways. For example, NOD1 and NOD2 activate NF-κB and MAPK pathways (25), whereas NLRP6, NLRC3, NLRC5, and NLRX1 have been demonstrated to regulate inflammation negatively (69).NLRP12 (NALP12, MONARCH-1, or PYPAF7) is a poorly characterized member of the NLR family. It has a tripartite domain structure, which consists of an N-terminal PYRIN domain, a central nucleotide binding site domain, and a C-terminal domain composed of at least 12 leucine-rich repeat motifs (10). In humans, NLRP12 is expressed in peripheral blood leukocytes, including granulocytes, eosinophils, monocytes, and dendritic cells (DCs) (10, 11). Similarly, mouse NLRP12 is highly expressed in bone marrow neutrophils and granulocytes, macrophages, and DCs (12, 13). Genetic studies in humans have shown that mutations in the NLRP12 gene are associated with periodic fever syndromes and atopic dermatitis (1416). More recent studies have demonstrated that NLRP12 has both inflammasome-dependent and inflammasome-independent roles in health and disease. Our laboratory and others have previously reported that NLRP12 mediates protection against colon inflammation and tumorigenesis in vivo by negatively regulating inflammatory responses (12, 17).Recent studies have revealed a potential role for NLRP12 during infectious diseases. Vladimer et al. (18) reported that Nlrp12−/− mice are hypersusceptible to Yersinia pestis infection, whereby NLRP12 is required to drive caspase-1 activation and IL-1β and IL-18 release. Another study found that WT and Nlrp12−/− mice exhibit similar host innate responses in lung infections induced by Mycobacterium tuberculosis or Klebsiella pneumoniae (13). However, in vitro studies reported that a synthetic analog cord factor, trehalose-6,6-dimycolate (TDP), from M. tuberculosis and LPS from K. pneumoniae induced substantially elevated levels of TNF-α and IL-6 in Nlrp12−/− bone marrow-derived DCs compared with their WT counterpart, although levels of secreted IL-1β were not changed (13). These results suggest that unlike the case in Yersinia infection, NLRP12 does not contribute to inflammasome-mediated protection against M. tuberculosis and K. pneumoniae infections. Overall, the physiological and functional relevance of NLRP12 in the host defense against infectious diseases is not fully understood.Salmonella enterica serovar Typhimurium (S. typhimurium) is a Gram-negative intracellular pathogen, and one of the most prevalent etiological agents of gastroenteritis worldwide. Salmonella infection accounts for 93.8 million cases of gastroenteritis annually in the world and is a leading cause of death among bacterial foodborne pathogens in the United States (19, 20). Previous studies have found that members of the Toll-like receptor (TLR) family, especially TLR4, are critical for the recognition and clearance of S. typhimurium (21, 22). One consequence of Salmonella-induced TLR activation is the production of inflammatory cytokines and antimicrobial compounds, including pro–IL-1β, pro–IL-18, IFN-γ, TNF-α, and reactive oxygen species, which are critical mediators for the control of bacterial growth in host tissues (23). In addition to TLR-mediated host responses, certain members of the NLR family, including NLRC4 and NLRP3, initiate inflammasome formation to drive processing and release of IL-1β and IL-18 following Salmonella infection (24, 25). Although the precise signals that trigger NLRP3 activation during Salmonella infection are unknown, NLRC4 is activated by NAIPs, a subset of receptors within the NLR family that detect Salmonella flagellin (mouse NAIP5 and NAIP6) or certain rod (mouse NAIP2) or needle (human NAIP and mouse NAIP1) proteins associated with the Salmonella type III secretion system (2630). Nevertheless, the functional relevance of NLRP12 in response to Salmonella infection is unknown.Here, we show that NLRP12 negatively regulates antibacterial host defense during Salmonella infection independent of inflammasomes. NLRP12 inhibited TLR-induced NF-κB activation by dampening phosphorylation of IκBα and ERK, consequently enhancing intracellular bacterial survival. Together, our work unveiled an NLRP12-dependent innate immune pathway that may be strategically exploited by S. typhimurium to persist and survive in the host.  相似文献   

2.
A series of mono- and dinuclear alkynylplatinum(II) terpyridine complexes containing the hydrophilic oligo(para-phenylene ethynylene) with two 3,6,9-trioxadec-1-yloxy chains was designed and synthesized. The mononuclear alkynylplatinum(II) terpyridine complex was found to display a very strong tendency toward the formation of supramolecular structures. Interestingly, additional end-capping with another platinum(II) terpyridine moiety of various steric bulk at the terminal alkyne would lead to the formation of nanotubes or helical ribbons. These desirable nanostructures were found to be governed by the steric bulk on the platinum(II) terpyridine moieties, which modulates the directional metal−metal interactions and controls the formation of nanotubes or helical ribbons. Detailed analysis of temperature-dependent UV-visible absorption spectra of the nanostructured tubular aggregates also provided insights into the assembly mechanism and showed the role of metal−metal interactions in the cooperative supramolecular polymerization of the amphiphilic platinum(II) complexes.Square-planar d8 platinum(II) polypyridine complexes have long been known to exhibit intriguing spectroscopic and luminescence properties (154) as well as interesting solid-state polymorphism associated with metal−metal and π−π stacking interactions (114, 25). Earlier work by our group showed the first example, to our knowledge, of an alkynylplatinum(II) terpyridine system [Pt(tpy)(C ≡ CR)]+ that incorporates σ-donating and solubilizing alkynyl ligands together with the formation of Pt···Pt interactions to exhibit notable color changes and luminescence enhancements on solvent composition change (25) and polyelectrolyte addition (26). This approach has provided access to the alkynylplatinum(II) terpyridine and other related cyclometalated platinum(II) complexes, with functionalities that can self-assemble into metallogels (2731), liquid crystals (32, 33), and other different molecular architectures, such as hairpin conformation (34), helices (3538), nanostructures (3945), and molecular tweezers (46, 47), as well as having a wide range of applications in molecular recognition (4852), biomolecular labeling (4852), and materials science (53, 54). Recently, metal-containing amphiphiles have also emerged as a building block for supramolecular architectures (4244, 5559). Their self-assembly has always been found to yield different molecular architectures with unprecedented complexity through the multiple noncovalent interactions on the introduction of external stimuli (4244, 5559).Helical architecture is one of the most exciting self-assembled morphologies because of the uniqueness for the functional and topological properties (6069). Helical ribbons composed of amphiphiles, such as diacetylenic lipids, glutamates, and peptide-based amphiphiles, are often precursors for the growth of tubular structures on an increase in the width or the merging of the edges of ribbons (64, 65). Recently, the optimization of nanotube formation vs. helical nanostructures has aroused considerable interests and can be achieved through a fine interplay of the influence on the amphiphilic property of molecules (66), choice of counteranions (67, 68), or pH values of the media (69), which would govern the self-assembly of molecules into desirable aggregates of helical ribbons or nanotube scaffolds. However, a precise control of supramolecular morphology between helical ribbons and nanotubes remains challenging, particularly for the polycyclic aromatics in the field of molecular assembly (6469). Oligo(para-phenylene ethynylene)s (OPEs) with solely π−π stacking interactions are well-recognized to self-assemble into supramolecular system of various nanostructures but rarely result in the formation of tubular scaffolds (7073). In view of the rich photophysical properties of square-planar d8 platinum(II) systems and their propensity toward formation of directional Pt···Pt interactions in distinctive morphologies (2731, 3945), it is anticipated that such directional and noncovalent metal−metal interactions might be capable of directing or dictating molecular ordering and alignment to give desirable nanostructures of helical ribbons or nanotubes in a precise and controllable manner.Herein, we report the design and synthesis of mono- and dinuclear alkynylplatinum(II) terpyridine complexes containing hydrophilic OPEs with two 3,6,9-trioxadec-1-yloxy chains. The mononuclear alkynylplatinum(II) terpyridine complex with amphiphilic property is found to show a strong tendency toward the formation of supramolecular structures on diffusion of diethyl ether in dichloromethane or dimethyl sulfoxide (DMSO) solution. Interestingly, additional end-capping with another platinum(II) terpyridine moiety of various steric bulk at the terminal alkyne would result in nanotubes or helical ribbons in the self-assembly process. To the best of our knowledge, this finding represents the first example of the utilization of the steric bulk of the moieties, which modulates the formation of directional metal−metal interactions to precisely control the formation of nanotubes or helical ribbons in the self-assembly process. Application of the nucleation–elongation model into this assembly process by UV-visible (UV-vis) absorption spectroscopic studies has elucidated the nature of the molecular self-assembly, and more importantly, it has revealed the role of metal−metal interactions in the formation of these two types of nanostructures.  相似文献   

3.
Studies in animal models and human subjects have shown that both innate and adaptive immunity contribute to the pathogenesis of type 1 diabetes (T1D). Whereas the role of TLR signaling pathways in T1D has been extensively studied, the contribution of the nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing protein (NLRP) 3 inflammasome pathway remains to be explored. In this study, we report that NLRP3 plays an important role in the development of T1D in the nonobese diabetic (NOD) mouse model. NLRP3 deficiency not only affected T-cell activation and Th1 differentiation, but also modulated pathogenic T-cell migration to the pancreatic islet. The presence of NLRP3 is critical for the expression of the chemokine receptors CCR5 and CXCR3 on T cells. More importantly, NLRP3 ablation reduced the expression of chemokine genes CCL5 and CXCL10 on pancreatic islet cells in an IRF-1–dependent manner. Our results suggest that molecules involved in chemotaxis, accompanied by the activation of the NLRP3 inflammasome, may be effective targets for the treatment of T1D.Type 1 diabetes (T1D) is a T-cell–mediated autoimmune disease characterized by the destruction of insulin-producing pancreatic beta cells in genetically predisposed individuals. Studies in animal models and human subjects have shown that both innate and adaptive immunity play a role in disease pathogenesis. Strategies targeting either T or B cells have shown some efficacy in T1D in both animal and human studies (14). Recently, the role of innate immunity in T1D has been increasingly appreciated. We, and others, have demonstrated that Toll-like receptor (TLR) signaling pathways are essential for the development of T1D. Nonobese diabetic (NOD) mice deficient in TLR2, TLR9, or MyD88 showed delayed disease development or were protected from diabetes (59). However, the development of autoimmune diabetes was accelerated in TLR4−/− NOD mice (57, 10). Whereas the role of TLR signaling has been intensively studied, the contribution of the nucleotide binding domain-like receptor (NLR) signaling pathway to the pathogenesis of T1D remains to be explored.Nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing protein (NLRP) 3 is a NLR family member, together with ASC and caspase-1, forms protein complexes that are responsible for the innate immune response to pathogens and/or “danger” signals (11). Increasing evidence indicates that the NLRP3 inflammasome plays an important role in obesity and type 2 diabetes (1214). However, little is known about the role of NLRP3 in autoimmune diabetes. Whereas the inflammasome has been extensively studied in the control of infection, only recently has the role of the NLRP inflammasome in autoimmune disease been recognized. Polymorphisms in inflammasome genes are involved in the predisposition to systemic lupus erythematosus (15). NLRP3 deficiency dramatically delayed the course and reduced severity of experimental autoimmune encephalomyelitis by suppression of Th1 and Th17 responses (16). Mice deficient in ASC, the adaptor protein of the NLRP3 inflammasome pathway, were also less susceptible to collagen-induced arthritis (17). Nevertheless, the role of the inflammasome pathway in the pathogenesis of T1D is unclear. Although caspase-1 or IL-1β deficiency did not protect NOD mice from T1D (18, 19), IL-1 blockade showed a synergistic protective effect when combined with anti-CD3 therapy for T1D in a mouse model (20). Interestingly, recent genetic association studies suggested that polymorphisms in inflammasome genes might be involved in the predisposition to T1D. A coding polymorphism in NLRP1 was demonstrated to confer susceptibility to T1D (21). Furthermore, two single-nucleotide polymorphisms in NLRP3 were identified in a separate association study as a predisposing factor for T1D (22).Thus, we generated NLRP3-deficient (NLRP3−/− or KO) NOD mice to understand the role of NLRP3 in the pathogenesis of T1D. Here, we show that NOD mice deficient in NLRP3 were protected from T1D development. Mechanistic studies suggested that the expression of NLRP3, in both hematopoietic and nonhematopoietic cells, was important for diabetes development. Whereas NLRP3 deficiency in the hematopoietic compartment reduced the diabetogenicity of immune cells, its ablation in nonhematopoietic cells, particularly in the pancreatic islets, compromised the migration of immune cells into the target tissue. Destruction of beta cells was reduced via the down-regulation of chemokine gene expression in the pancreatic islets leading to protection from diabetes.  相似文献   

4.
5.
Pathogen recognition by nucleotide-binding oligomerization domain-like receptor (NLR) results in the formation of a macromolecular protein complex (inflammasome) that drives protective inflammatory responses in the host. It is thought that the number of inflammasome complexes forming in a cell is determined by the number of NLRs being activated, with each NLR initiating its own inflammasome assembly independent of one another; however, we show here that the important foodborne pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) simultaneously activates at least two NLRs, whereas only a single inflammasome complex is formed in a macrophage. Both nucleotide-binding domain and leucine-rich repeat caspase recruitment domain 4 and nucleotide-binding domain and leucine-rich repeat pyrin domain 3 are simultaneously present in the same inflammasome, where both NLRs are required to drive IL-1β processing within the Salmonella-infected cell and to regulate the bacterial burden in mice. Superresolution imaging of Salmonella-infected macrophages revealed a macromolecular complex with an outer ring of apoptosis-associated speck-like protein containing a caspase activation and recruitment domain and an inner ring of NLRs, with active caspase effectors containing the pro–IL-1β substrate localized internal to the ring structure. Our data reveal the spatial localization of different components of the inflammasome and how different members of the NLR family cooperate to drive robust IL-1β processing during Salmonella infection.Inflammasomes are cytosolic multimeric protein complexes formed in the host cell in response to the detection of pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs). Formation of the inflammasome in response to PAMPs is critical for host defense because it facilitates processing of the proinflammatory cytokines pro–IL-1β and pro–IL-18 into their mature forms (1). The inflammasome also initiates host cell death in the form of pyroptosis, releasing macrophage-resident microbes to be killed by other immune mechanisms (2). The current paradigm is that there are individual, receptor-specific inflammasomes consisting of one nucleotide-binding oligomerization domain-like receptor (NLR; leucine-rich repeat–containing) or PYHIN [pyrin domain and hematopoietic expression, interferon-inducible nature, and nuclear localization (HIN) domain-containing] receptor, the adaptor protein apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (CARD; ASC), and caspase-1 (3). How the protein constituents of the inflammasome are spatially orientated is unclear.Nucleotide-binding domain and leucine-rich repeat caspase recruitment domain 4 (NLRC4) and nucleotide-binding domain and leucine-rich repeat pyrin domain 3 (NLRP3) are the best-characterized inflammasomes, especially with respect to their responses to pathogenic bacteria. The NLRC4 inflammasome is activated primarily by bacteria, including Aeromonas veronii (4), Escherichia coli (5), Listeria monocytogenes (6, 7), Pseudomonas aeruginosa (5), Salmonella enterica serovar Typhimurium (S. Typhimurium) (5, 810), and Yersinia species (11). In mouse macrophages, the NLRC4 inflammasome responds to flagellin and type III secretion system-associated needle or rod proteins (5, 8, 9) after their detection by NLR family, apoptosis inhibitory protein (NAIP) 5 or NAIP6 and NAIP1 or NAIP2, respectively (1215). Phosphorylation of NLRC4 at a single, evolutionarily conserved residue, Ser 533, by PKCδ kinase is required for NLRC4 inflammasome assembly (16). The NLRP3 inflammasome is activated by a large repertoire of DAMPs, including ATP, nigericin, maitotoxin, uric acid crystals, silica, aluminum hydroxide, and muramyl dipeptide (1720). NLRP3 is also activated by bacterial PAMPs from many species, including Aeromonas species (4, 21), L. monocytogenes (6, 7, 22), Neisseria gonorrhoeae (23), S. Typhimurium (10), Streptococcus pneumoniae (24), and Yersinia species (11). The mechanisms by which NLRC4 and NLRP3 inflammasomes contribute to host defense against bacterial pathogens are emerging; however, little is known about the dynamics governing inflammasome assembly in infections caused by bacteria that activate multiple NLRs, such as S. Typhimurium (10), A. veronii (4), and Yersinia (11).NLRP3 does not have a CARD and requires ASC to interact with the CARD of procaspase-1. This interaction requires a charged interface around Asp27 of the procaspase-1 CARD (25). Whether ASC is also required for the assembly of the NLRC4 inflammasome is less clear. NLRC4 contains a CARD that can interact directly with the CARD of procaspase-1 (26); however, ASC is required for some of the responses driven by NLRC4 (27). Macrophages infected with S. Typhimurium or other pathogens exhibit formation of a distinct cytoplasmic ASC focus or speck, which can be visualized under the microscope and is indicative of inflammasome activation (10, 28, 29). Our laboratory and others have shown that only one ASC speck is formed per cell irrespective of the stimulus used (2932). However, many bacteria activate two or more NLRs, and it is unclear whether a singular inflammasome is formed at a time or if multiple inflammasomes are formed independent of each other, with each inflammasome containing one member of the NLR family.In this study, we describe the endogenous molecular constituents of the Salmonella-induced inflammasome and their spatial orientation. In cross-section, ASC forms a large external ring with the NLRs and caspases located internally. Critically, NLRC4, NLRP3, caspase-1, and caspase-8 coexist in the same ASC speck to coordinate pro–IL-1β processing. All ASC specks observed contained both NLRC4 and NLRP3. These results suggest that Salmonella infection induces a single inflammasome protein complex containing different NLRs and recruiting multiple caspases to coordinate a multifaceted inflammatory response to infection.  相似文献   

6.
It was recently demonstrated that in ferric myoglobins (Mb) the fluorescence quenching of the photoexcited tryptophan 14 (*Trp14) residue is in part due to an electron transfer to the heme porphyrin (porph), turning it to the ferrous state. However, the invariance of *Trp decay times in ferric and ferrous Mbs raises the question as to whether electron transfer may also be operative in the latter. Using UV pump/visible probe transient absorption, we show that this is indeed the case for deoxy-Mb. We observe that the reduction generates (with a yield of about 30%) a low-valence Fe–porphyrin π [FeII(porph●−)] -anion radical, which we observe for the first time to our knowledge under physiological conditions. We suggest that the pathway for the electron transfer proceeds via the leucine 69 (Leu69) and valine 68 (Val68) residues. The results on ferric Mbs and the present ones highlight the generality of Trp–porphyrin electron transfer in heme proteins.Electron transfer plays a fundamental role in many biological systems (13) ranging from photosynthetic proteins (4) to iron–sulfur (5), copper (6), and heme (7, 8) proteins. It was demonstrated that electron transfer can be used to produce from heme proteins in situ drugs with antimalarial activity (9) and it might have a role in protein folding (2). In general, electron transfer in proteins can occur over long distances (>10 Å) by hopping through different residues, thus reducing the time that would be needed for a single step tunneling from the donor to the acceptor (1012). Aromatic amino acids and Tryptophan (Trp) in particular can act as a relay in such processes (1319). Trp also acts as a phototriggered electron donor, e.g., in DNA repair by photolyase (1618) and in cryptochromes (20, 21). When no obvious electron acceptors are present, excited Trp or (*Trp) still displays shorter lifetimes than its nanosecond decay times in solution (22, 23). This is due to its strong tendency to act as an electron donor, undergoing electron transfer toward the protein’s backbone as in the case of apo-myoglobin mutants (24), small cyclic peptides (25), and human γ–d-crystallin (26). It is interesting to note that in wild-type horse heart (WT-HH) apo-myoglobin the fluorescence lifetime of the two *Trp residues was reported to be comparable to that in water (27), demonstrating the absence of deactivation mechanisms, either by energy or by electron transfer.The protein visible absorption spectrum is dominated by their cofactors, e.g., heme or flavins, whereas the UV absorption in the region between 250 nm and 300 nm is mainly due to the three aromatic amino acids, Trp, tyrosine (Tyr), and phenylalanine (Phe) (28), with Trp having the highest molar extinction coefficient. The high sensitivity of Trp to the local environment and the possibility to correlate it with its fluorescence response (28) have led to its widespread use as a local natural probe of protein structure and dynamics in time-resolved fluorescence resonance energy transfer (FRET) studies, and it has emerged as the “spectroscopic ruler” in such studies (2830). FRET is mediated by dipole–dipole coupling between a donor *Trp and an acceptor molecule, and its rate is inversely proportional to the sixth power of the distance between them and to the relative orientation of their dipoles.Myoglobin (Mb) is a small heme protein composed of ∼150 residues (31) arranged in eight α-helices (from A to H) (SI Appendix, Fig. S7), whose biological function is to store molecular oxygen in muscles of vertebrates (32). This is accomplished by its prosthetic group: a Fe–Protoporphyrin IX complex bound to the protein structure via the proximal histidine (His93) (SI Appendix, Fig. S7). Both ferric and ferrous hemes tend to bind small diatomic molecules (e.g., O2, CO, NO, and CN) at the Fe site. Mb has two Trp residues that are situated in the α-helix A: Trp7 toward the solvent and Trp14 within the protein and closer to the heme (SI Appendix, Fig. S7) (33). Previous time-resolved fluorescence studies on various Mb complexes have reported decay times (SI Appendix, Table S1) of ∼120 ps and ∼20 ps, for *Trp7 and *Trp14, respectively (3438). These decay times appear invariant with respect to the ligand and the oxidation state of the iron ion in the heme. They were attributed to *Trp-to-porphyrin energy transfer via FRET over different donor–acceptor distances (37, 38) [the Trp7-Heme and Trp14-Heme center-to-center distances are 21.2 Å and 15.1 Å, respectively (33, 39) (SI Appendix, Fig. S7)]. We recently showed, using ultrafast 2D-UV and visible transient absorption (TA) spectroscopy, that in the ferric myoglobins (MbCN and MbH2O) the relaxation pathway of *Trp14 involves not only a *Trp-to-heme FRET but also an electron transfer from the *Trp to the heme (40) in a ratio of approximately 60–40%. One can expect that due to its ferric character, the heme is a strong electron acceptor in these cases, and indeed our study showed the formation of an FeII heme.However, the invariance of *Trp decay times in ferric and ferrous Mbs (SI Appendix, Table S1) suggests that similar electron transfer processes may also occur in ferrous Mbs. In this event, questions arise as to (i) whether a formally FeI heme is formed, which has to date been observed only in cryo-radiolysis experiments (41, 42), or (ii) whether the electron localizes on the porphyrin ring or even on the ligand that binds to the Fe ion. Theoretical investigations have suggested that an iron porphyrin anion radical can be formed (4345).To address these questions, here we present a UV-pump/visible-probe TA study of ferrous Mbs. In the latter case with apical diatomic ligands, e.g., MbNO and MbCO, heme photoexcitation leads to dissociation of the ligand, followed by its recombination to the heme, which can be both geminate (the ligand stays inside the protein scaffold) and nongeminate (the ligand migrates out of the protein scaffold) (4648). For the NO ligand, recombination timescales are typically ∼10 ps, ∼30 ps, and ∼200 ps (46, 47), whereas for CO they span up to the millisecond range (46, 4951). The presence of recombination timescales in the order of *Trp decay times leads to additional signal contributions, which complicate the analysis of the data. These problems are avoided using deoxy-Mb, which has a penta-coordinated heme bound only to the His93. Upon heme photoexcitation, the system recovers to the ground state within a few picoseconds (46, 52). This allows investigating the *Trp–heme interaction without any overlapping contributions.We show here that just as in the ferric Mbs (40), also in deoxy-Mb does *Trp14 partly decay to the heme by electron transfer, competing with the FRET pathway. We find that the transferred electron is localized on the porphyrin ring, contrary to the ferric case where it resides on the metal center. This is due to the highly negative reduction potential of the FeII/FeI couple (53, 54), which is close to the porphyrin reduction potential (55). To our knowledge, this is the first report of a low-valent myoglobin, under physiological conditions.The experimental setup, the sample preparation, and the data analysis are described in SI Appendix.  相似文献   

7.
8.
Nod-like receptor family, pyrin domain-containing 3 (NLRP3), is involved in the early stages of the inflammatory response by sensing cellular damage or distress due to viral or bacterial infection. Activation of NLRP3 triggers its assembly into a multimolecular protein complex, termed “NLRP3 inflammasome.” This event leads to the activation of the downstream molecule caspase-1 that cleaves the precursor forms of proinflammatory cytokines, such as interleukin 1 beta (IL-1β) and IL-18, and initiates the immune response. Recent studies indicate that the reactive oxygen species produced by mitochondrial respiration is critical for the activation of the NLRP3 inflammasome by monosodium urate, alum, and ATP. However, the precise mechanism by which RNA viruses activate the NLRP3 inflammasome is not well understood. Here, we show that loss of mitochondrial membrane potential [ΔΨ(m)] dramatically reduced IL-1β secretion after infection with influenza, measles, or encephalomyocarditis virus (EMCV). Reduced IL-1β secretion was also observed following overexpression of the mitochondrial inner membrane protein, uncoupling protein-2, which induces mitochondrial proton leakage and dissipates ΔΨ(m). ΔΨ(m) was required for association between the NLRP3 and mitofusin 2, a mediator of mitochondrial fusion, after infection with influenza virus or EMCV. Importantly, the knockdown of mitofusin 2 significantly reduced the secretion of IL-1β after infection with influenza virus or EMCV. Our results provide insight into the roles of mitochondria in NLRP3 inflammasome activation.Nod-like receptor family, pyrin domain-containing 3 (NLRP3) can be activated by a wide variety of stimuli, such as endogenous danger signals from damaged cells, bacterial components, environmental irritants, and DNA and RNA viruses (1). It forms a multiprotein complex called the NLRP3 inflammasome, which includes an adaptor protein apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and procaspase-1. The NLRP3 inflammasome-mediated cytokine release requires two signaling pathways (2). The first signal is induced by Toll-like receptors (TLRs), interleukin 1 receptor (IL-1R), or tumor necrosis factor receptor, and leads to the synthesis of inactive NLRP3, pro–IL-1β, and pro–IL-18 in the cytosol. The second signal is triggered by NLRP3 agonists, which induce the activation of caspase-1. Caspase-1 catalyzes the proteolytic processing of pro–IL-1β and pro–IL-18, and their conversion to mature forms, and stimulates their secretion across the plasma membrane (1). These inflammasome-dependent cytokines play a key role in the induction of adaptive immunity and the initiation of tissue healing after influenza virus infection (35). Migration of dendritic cells (DCs) to the draining lymph nodes and priming of CD8 T cells during influenza virus infection require IL-1R signaling in respiratory DCs (6). By contrast, chronic activation of the NLRP3 inflammasome has been linked to many inflammatory diseases (7, 8). Therefore, increasing the number of studies dedicated to the investigation of the molecular mechanisms of NLRP3 inflammasome activation will be crucial for improving our understanding of the pathogenesis of infectious and autoinflammatory diseases.Mitochondria are compartmentalized by two membrane bilayers (outer and inner membranes) and are involved in a wide variety of functions in eukaryotic cells. Within the past decade, novel functions of mitochondria have been discovered demonstrating their crucial role in innate antiviral immunity in vertebrates (9). A direct link between mitochondria and innate immunity was first highlighted with the finding that an adaptor protein, mitochondrial antiviral signaling (MAVS; also known as IPS-1, VISA, or Cardif) (1013), triggered retinoic acid-inducible gene 1 (RIG-I) and melanoma differentiation-associated protein 5-mediated type I interferon (IFN) induction. In addition to their role in antiviral immunity, mitochondria also function as a platform for the activation of the NLRP3 inflammasome by producing mitochondrial reactive oxygen species (mROS) (14, 15). In this context, NLRP3 agonists trigger the generation of mROS from damaged mitochondria, resulting in the dissociation of thioredoxin (TRX) from TRX-interacting protein, which associates with NLRP3 to facilitate inflammasome formation (16). Furthermore, cytosolic mitochondrial DNA (mtDNA) released from damaged mitochondria has been reported to activate the NLRP3 inflammasome (17) and absent in melanoma 2 inflammasome (15), recently identified as a cytoplasmic DNA sensor for the inflammasome (1821). Although mitochondria are essential for host-cell defense, the mechanism of their involvement in the activation of the NLRP3 inflammasome is still unclear. In the present study, we demonstrate that the mitofusin 2 (Mfn2) is required for the full activation of the NLRP3 inflammasomes in macrophages.  相似文献   

9.
Protein toxins from tarantula venom alter the activity of diverse ion channel proteins, including voltage, stretch, and ligand-activated cation channels. Although tarantula toxins have been shown to partition into membranes, and the membrane is thought to play an important role in their activity, the structural interactions between these toxins and lipid membranes are poorly understood. Here, we use solid-state NMR and neutron diffraction to investigate the interactions between a voltage sensor toxin (VSTx1) and lipid membranes, with the goal of localizing the toxin in the membrane and determining its influence on membrane structure. Our results demonstrate that VSTx1 localizes to the headgroup region of lipid membranes and produces a thinning of the bilayer. The toxin orients such that many basic residues are in the aqueous phase, all three Trp residues adopt interfacial positions, and several hydrophobic residues are within the membrane interior. One remarkable feature of this preferred orientation is that the surface of the toxin that mediates binding to voltage sensors is ideally positioned within the lipid bilayer to favor complex formation between the toxin and the voltage sensor.Protein toxins from venomous organisms have been invaluable tools for studying the ion channel proteins they target. For example, in the case of voltage-activated potassium (Kv) channels, pore-blocking scorpion toxins were used to identify the pore-forming region of the channel (1, 2), and gating modifier tarantula toxins that bind to S1–S4 voltage-sensing domains have helped to identify structural motifs that move at the protein–lipid interface (35). In many instances, these toxin–channel interactions are highly specific, allowing them to be used in target validation and drug development (68).Tarantula toxins are a particularly interesting class of protein toxins that have been found to target all three families of voltage-activated cation channels (3, 912), stretch-activated cation channels (1315), as well as ligand-gated ion channels as diverse as acid-sensing ion channels (ASIC) (1621) and transient receptor potential (TRP) channels (22, 23). The tarantula toxins targeting these ion channels belong to the inhibitor cystine knot (ICK) family of venom toxins that are stabilized by three disulfide bonds at the core of the molecule (16, 17, 2431). Although conventional tarantula toxins vary in length from 30 to 40 aa and contain one ICK motif, the recently discovered double-knot toxin (DkTx) that specifically targets TRPV1 channels contains two separable lobes, each containing its own ICK motif (22, 23).One unifying feature of all tarantula toxins studied thus far is that they act on ion channels by modifying the gating properties of the channel. The best studied of these are the tarantula toxins targeting voltage-activated cation channels, where the toxins bind to the S3b–S4 voltage sensor paddle motif (5, 3236), a helix-turn-helix motif within S1–S4 voltage-sensing domains that moves in response to changes in membrane voltage (3741). Toxins binding to S3b–S4 motifs can influence voltage sensor activation, opening and closing of the pore, or the process of inactivation (4, 5, 36, 4246). The tarantula toxin PcTx1 can promote opening of ASIC channels at neutral pH (16, 18), and DkTx opens TRPV1 in the absence of other stimuli (22, 23), suggesting that these toxin stabilize open states of their target channels.For many of these tarantula toxins, the lipid membrane plays a key role in the mechanism of inhibition. Strong membrane partitioning has been demonstrated for a range of toxins targeting S1–S4 domains in voltage-activated channels (27, 44, 4750), and for GsMTx4 (14, 50), a tarantula toxin that inhibits opening of stretch-activated cation channels in astrocytes, as well as the cloned stretch-activated Piezo1 channel (13, 15). In experiments on stretch-activated channels, both the d- and l-enantiomers of GsMTx4 are active (14, 50), implying that the toxin may not bind directly to the channel. In addition, both forms of the toxin alter the conductance and lifetimes of gramicidin channels (14), suggesting that the toxin inhibits stretch-activated channels by perturbing the interface between the membrane and the channel. In the case of Kv channels, the S1–S4 domains are embedded in the lipid bilayer and interact intimately with lipids (48, 51, 52) and modification in the lipid composition can dramatically alter gating of the channel (48, 5356). In one study on the gating of the Kv2.1/Kv1.2 paddle chimera (53), the tarantula toxin VSTx1 was proposed to inhibit Kv channels by modifying the forces acting between the channel and the membrane. Although these studies implicate a key role for the membrane in the activity of Kv and stretch-activated channels, and for the action of tarantula toxins, the influence of the toxin on membrane structure and dynamics have not been directly examined. The goal of the present study was to localize a tarantula toxin in membranes using structural approaches and to investigate the influence of the toxin on the structure of the lipid bilayer.  相似文献   

10.
Inflammasomes are caspase-1–activating multiprotein complexes. The mouse nucleotide-binding domain and leucine rich repeat pyrin containing 1b (NLRP1b) inflammasome was identified as the sensor of Bacillus anthracis lethal toxin (LT) in mouse macrophages from sensitive strains such as BALB/c. Upon exposure to LT, the NLRP1b inflammasome activates caspase-1 to produce mature IL-1β and induce pyroptosis. Both processes are believed to depend on autoproteolysed caspase-1. In contrast to human NLRP1, mouse NLRP1b lacks an N-terminal pyrin domain (PYD), indicating that the assembly of the NLRP1b inflammasome does not require the adaptor apoptosis-associated speck-like protein containing a CARD (ASC). LT-induced NLRP1b inflammasome activation was shown to be impaired upon inhibition of potassium efflux, which is known to play a major role in NLRP3 inflammasome formation and ASC dimerization. We investigated whether NLRP3 and/or ASC were required for caspase-1 activation upon LT stimulation in the BALB/c background. The NLRP1b inflammasome activation was assessed in both macrophages and dendritic cells lacking either ASC or NLRP3. Upon LT treatment, the absence of NLRP3 did not alter the NLRP1b inflammasome activity. Surprisingly, the absence of ASC resulted in IL-1β cleavage and pyroptosis, despite the absence of caspase-1 autoprocessing activity. By reconstituting caspase-1/caspase-11−/− cells with a noncleavable or catalytically inactive mutant version of caspase-1, we directly demonstrated that noncleavable caspase-1 is fully active in response to the NLRP1b activator LT, whereas it is nonfunctional in response to the NLRP3 activator nigericin. Taken together, these results establish variable requirements for caspase-1 cleavage depending on the pathogen and the responding NLR.Anthrax is a zoonotic disease caused by the Gram-positive bacterium Bacillus anthracis. B. anthracis provokes a shock-like syndrome that can prove fatal to the host (1) and has recently gained notoriety as a potential bioterrorism agent. Anthrax pathogenicity relies on its ability to secrete three virulence proteins, which combine with each other to form two toxins. The protective antigen (PA) combines with the edema factor (EF) to form the edema toxin (2, 3). EF is an adenylate cyclase that causes edema of the infected tissue. The binary combination of PA with lethal factor (LF) gives rise to the most virulent factor, called lethal toxin (LT), responsible for the systemic symptoms and death of the infected animal. To escape the host immune response, LT impairs the host innate immunity by killing macrophages (46). The PA protein interacts with LF and binds to cell surface receptors, enabling endocytosis of the LT complex. In the acidic compartment, PA forms pores allowing the delivery of LF to the cytosol. LF is a zinc metalloprotease that was shown to cleave the N-terminal region of many MAP kinase kinases and to induce apoptosis of macrophages. LT also triggers pyroptosis through the formation of a caspase-1–activating platform, named “inflammasome” (68).Inflammasomes are multiprotein complexes of the innate immune response that control caspase-1 activity and pro–IL-1β and pro–IL-18 maturation. Most inflammasomes are composed of specific cytosolic pathogen recognition receptors (PRRs), as well as the apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (CARD) (ASC) adaptor protein that enables the recruitment and activation of the caspase-1 protease. Once caspase-1 is oligomerized within an inflammasome platform, the enzyme undergoes autoproteolysis to form heterodimers of active caspase-1 (912). In the mouse, at least five distinct inflammasomes have been described, distinguished by the PRR that induces the complex formation. The PRRs capable of participating in inflammasome platform formation are either members of the nod-like receptor (NLR) family (e.g., NLRP1, NLRP3, or NLRC4) or of the PYrin and HIN (PYHIN) family (e.g., AIM2) (13, 14). ASC is composed of a pyrin domain (PYD) and a caspase activation and recruitment domain (CARD). ASC interacts with a PYD-containing PRR via its PYD domain and recruits the CARD domain of caspase-1 via its CARD domain. Thus, ASC is essential to the formation of the inflammasome by receptors such as NLRP3 or AIM2 (1518). However, its presence is dispensable for NLRC4, which contains a CARD in place of a PYD, allowing direct interaction with the CARD domain of caspase-1 (19, 20).Past studies have determined that certain mouse strains are more sensitive than others to LT cytotoxicity, and genetic studies identified NLRP1b as the factor conferring mouse strain susceptibility to anthrax LT (21). The mouse genome contains three different NLRP1 isoforms (a, b, and c) and a functional NLRP1b was found to be expressed by the mouse strains sensitive to LT (e.g., BALB/c or 129 background). Expression of NLRP1b was shown to mediate IL-1β release and caspase-1–mediated cell death in response to LT (7, 21, 22). Mouse NLRP1b differs structurally from human NLRP1 in that it lacks the N-terminal PYD (23). The absence of the PYD suggests that NLRP1b can directly engage caspase-1 without a requirement for ASC. However, studies dissecting the mechanism of NLRC4 inflammasome activation demonstrated that ASC is required for the amplification of caspase-1 autoprocessing and IL-1β secretion but not for pyroptosis (19, 20). Cell lysis mediated by LT was shown to be dependent on sodium and potassium fluxes (24), and high extracellular potassium inhibited IL-1β secretion upon LT treatment, suggesting a role for the NLRP3 inflammasome in LT sensing (22, 25). Therefore, we investigated whether NLRP3 and/or ASC were required for caspase-1 activation in response to LT. The NLRP3, ASC, and caspase-1 mouse knockout strains were backcrossed into the BALB/c background and the response of macrophages and dendritic cells (DCs) to LT intoxication was studied. Our data reveal that (i) in response to LT, ASC is dispensable for caspase-1 activation, but uncleavable caspase-1 is fully active; and (ii) upon activation of the NLRP3 inflammasome, uncleavable caspase-1 is inactive.  相似文献   

11.
Background and objectives: Natriuretic peptides have been suggested to be of value in risk stratification in dialysis patients. Data in patients on peritoneal dialysis remain limited.Design, setting, participants, & measurements: Patients of the ADEMEX trial (ADEquacy of peritoneal dialysis in MEXico) were randomized to a control group [standard 4 × 2L continuous ambulatory peritoneal dialysis (CAPD); n = 484] and an intervention group (CAPD with a target creatinine clearance ≥60L/wk/1.73 m2; n = 481). Natriuretic peptides were measured at baseline and correlated with other parameters as well as evaluated for effects on patient outcomes.Results: Control group and intervention group were comparable at baseline with respect to all measured parameters. Baseline values of natriuretic peptides were elevated and correlated significantly with levels of residual renal function but not with body size or diabetes. Baseline values of N-terminal fragment of B-type natriuretic peptide (NT-proBNP) but not proANP(1–30), proANP(31–67), or proANP(1–98) were independently highly predictive of overall survival and cardiovascular mortality. Volume removal was also significantly correlated with patient survival.Conclusions. NT-proBNP have a significant predictive value for survival of CAPD patients and may be of value in guiding risk stratification and potentially targeted therapeutic interventions.Plasma levels of cardiac natriuretic peptides are elevated in patients with chronic kidney disease, owing to impairment of renal function, hypertension, hypervolemia, and/or concomitant heart disease (17). Atrial natriuretic peptide (ANP) and particularly brain natriuretic peptide (BNP) levels are linked independently to left ventricular mass (35,816) and function (3,617) and predict total and cardiovascular mortality (1,3,8,10,12,18) as well as cardiac events (12,19). ANP and BNP decrease significantly during hemodialysis treatment but increase again during the interdialytic interval (1,2,4,6,7,14,17,2023). Levels in patients on peritoneal dialysis (PD) have been found to be lower than in patients on hemodialysis (11,2426), but the correlations with left ventricular function and structure are maintained in both types of dialysis modalities (11,15,27,28).The high mortality of patients on peritoneal dialysis and the failure of dialytic interventions to alter this mortality (29,30) necessitate renewed attention into novel methods of stratification and identification of patients at highest risk to be targeted for specific interventions. Cardiac natriuretic peptides are increasingly considered to fulfill this role in nonrenal patients. Evaluations of cardiac natriuretic peptides in patients on PD have been limited by small numbers (3,9,11,12,15,2426) and only one study examined correlations between natriuretic peptide levels and outcomes (12). The PD population enrolled in the ADEMEX trial offered us the opportunity to evaluate cardiac natriuretic peptides and their value in predicting outcomes in the largest clinical trial ever performed on PD (29,30). It is hoped that such an evaluation would identify patients at risk even in the absence of overt clinical disease and hence facilitate or encourage interventions with salutary outcomes.  相似文献   

12.
13.
Antiretroviral therapy (ART) reduces the infectiousness of HIV-infected persons, but only after testing, linkage to care, and successful viral suppression. Thus, a large proportion of HIV transmission during a period of high infectiousness in the first few months after infection (“early transmission”) is perceived as a threat to the impact of HIV “treatment-as-prevention” strategies. We created a mathematical model of a heterosexual HIV epidemic to investigate how the proportion of early transmission affects the impact of ART on reducing HIV incidence. The model includes stages of HIV infection, flexible sexual mixing, and changes in risk behavior over the epidemic. The model was calibrated to HIV prevalence data from South Africa using a Bayesian framework. Immediately after ART was introduced, more early transmission was associated with a smaller reduction in HIV incidence rate—consistent with the concern that a large amount of early transmission reduces the impact of treatment on incidence. However, the proportion of early transmission was not strongly related to the long-term reduction in incidence. This was because more early transmission resulted in a shorter generation time, in which case lower values for the basic reproductive number (R0) are consistent with observed epidemic growth, and R0 was negatively correlated with long-term intervention impact. The fraction of early transmission depends on biological factors, behavioral patterns, and epidemic stage and alone does not predict long-term intervention impacts. However, early transmission may be an important determinant in the outcome of short-term trials and evaluation of programs.Recent studies have confirmed that effective antiretroviral therapy (ART) reduces the transmission of HIV among stable heterosexual couples (13). This finding has generated interest in understanding the population-level impact of HIV treatment on reducing the rate of new HIV infections in generalized epidemic settings (4). Research, including mathematical modeling (510), implementation research (11), and major randomized controlled trials (1214), are focused on how ART provision might be expanded strategically to maximize its public health benefits (15, 16).One concern is that if a large fraction of HIV transmission occurs shortly after a person becomes infected, before the person can be diagnosed and initiated on ART, this will limit the potential impact of HIV treatment on reducing HIV incidence (9, 17, 18). Data suggest that persons are more infectious during a short period of “early infection” after becoming infected with HIV (1922), although there is debate about the extent, duration, and determinants of elevated infectiousness (18, 23). The amount of transmission that occurs also will depend on patterns of sexual behavior and sexual networks (17, 2427). There have been estimates for the contribution of early infection to transmission from mathematical models (7, 17, 21, 2426) and phylogenetic analyses (2831), but these vary widely, from 5% to above 50% (23).In this study, we use a mathematical model to quantify how the proportion of transmission that comes from persons who have been infected recently affects the impact of treatment scale-up on HIV incidence. The model is calibrated to longitudinal HIV prevalence data from South Africa using a Bayesian framework. Thus, the model accounts for not only the early epidemic growth rate highlighted in previous research (5, 9, 18), but also the heterogeneity and sexual behavior change to explain the peak and decline in HIV incidence observed in sub-Saharan African HIV epidemics (32, 33).The model calibration allows uncertainty about factors that determine the amount of early transmission, including the relative infectiousness during early infection, heterogeneity in propensity for sexual risk behavior, assortativity in sexual partner selection, reduction in risk propensity over the life course, and population-wide reductions in risk behavior in response to the epidemic (32, 33). This results in multiple combinations of parameter values that are consistent with the observed epidemic and variation in the amount of early transmission. We simulated the impact of a treatment intervention and report how the proportion of early transmission correlates with the reduction in HIV incidence from the intervention over the short- and long-term.  相似文献   

14.
Inflammasomes are intracellular sensors that couple detection of pathogens and cellular stress to activation of Caspase-1, and consequent IL-1β and IL-18 maturation and pyroptotic cell death. Here, we show that the absent in melanoma 2 (AIM2) and nucleotide-binding oligomerization domain-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasomes trigger Caspase-1–dependent mitochondrial damage. Caspase-1 activates multiple pathways to precipitate mitochondrial disassembly, resulting in mitochondrial reactive oxygen species (ROS) production, dissipation of mitochondrial membrane potential, mitochondrial permeabilization, and fragmentation of the mitochondrial network. Moreover, Caspase-1 inhibits mitophagy to amplify mitochondrial damage, mediated in part by cleavage of the key mitophagy regulator Parkin. In the absence of Parkin activity, increased mitochondrial damage augments pyroptosis, as indicated by enhanced plasma membrane permeabilization and release of danger-associated molecular patterns (DAMPs). Therefore, like other initiator caspases, Caspase-1 activation by inflammasomes results in mitochondrial damage.Inflammasomes are cytosolic complexes that mediate Caspase-1 activation in response to pathogen infection and cellular stress (1, 2). They consist of a regulatory subunit, which couples stimulus recognition to complex assembly; Caspase-1, the effector subunit; and the adaptor protein Asc. The best-characterized inflammasomes include the AIM2 inflammasome, which detects cytosolic DNA during bacterial and viral infection, and the NLRP3 inflammasome, which is activated by many stimuli in a variety of settings including infection and metabolic inflammation. Although not entirely clear, one plausible model of NLRP3 inflammasome activation is the generation of some mitochondria-associated signal by mitochondrial destabilization (3, 4). Recruitment of Caspase-1 into the inflammasome complex leads to its activation, autoprocessing, and subsequent substrate cleavage.The prototypical inflammasome-mediated functions are IL-1β and IL-18 maturation and induction of pyroptosis (1). Additionally, inflammasomes control other processes like unconventional secretion of intracellular proteins (5), such as DAMPs like high mobility group box 1 (HMGB1) (6), and regulation of autophagy (7, 8). These examples suggest the existence of additional inflammasome effector activities that are likely to vary in a context-dependent manner. Interestingly, a recent report indicated that activation of the NLRP3 inflammasome by extracellular ATP leads to NLRP3-dependent dissipation of the mitochondrial membrane potential (9), but subsequent studies proposed that such mitochondrial damage is solely a trigger of inflammasome activation (10, 11) because it occurs normally in the absence of the NLRP3 inflammasome (11). Thus, the relationship between NLRP3 inflammasome activation and mitochondrial damage remains unclear.Pyroptosis is a Caspase-1–mediated, proinflammatory form of cell death. It occurs during infection by many intracellular pathogens where it can critically eliminate an intracellular replication niche (12), as well as other settings (13, 14), but experimental demonstration of its physiological role is hampered by the lack of mechanistic insights into its regulation. Pyroptosis shares some features with necrosis (such as loss of plasma-membrane integrity and release of intracellular contents) and others with apoptosis (including DNA fragmentation and nuclear condensation) (12). Mitochondrial damage critically underlies apoptosis mediated by initiator caspases like Caspase-8 and Caspase-9. Upon activation by death receptors, Caspase-8 cleavage of the protein Bid precipitates mitochondrial outer membrane permeabilization (MOMP), resulting in dissipation of the membrane potential, disruption of mitochondrial function, and release of apoptosis-promoting factors from the intermembrane space (15). MOMP can also lead to mitochondrial permeability transition (MPT), or breach of integrity of the inner membrane caused by opening of an inner membrane pore, which further amplifies mitochondrial damage (16). Whether mitochondrial damage contributes to pyroptosis and in general how pyroptosis is regulated, including the relevant substrates, are not clear.  相似文献   

15.
Inflammation that contributes to acute cerebrovascular disease is driven by the proinflammatory cytokine interleukin-1 and is known to exacerbate resulting injury. The activity of interleukin-1 is regulated by multimolecular protein complexes called inflammasomes. There are multiple potential inflammasomes activated in diverse diseases, yet the nature of the inflammasomes involved in brain injury is currently unknown. Here, using a rodent model of stroke, we show that the NLRC4 (NLR family, CARD domain containing 4) and AIM2 (absent in melanoma 2) inflammasomes contribute to brain injury. We also show that acute ischemic brain injury is regulated by mechanisms that require ASC (apoptosis-associated speck-like protein containing a CARD), a common adaptor protein for several inflammasomes, and that the NLRP3 (NLR family, pyrin domain containing 3) inflammasome is not involved in this process. These discoveries identify the NLRC4 and AIM2 inflammasomes as potential therapeutic targets for stroke and provide new insights into how the inflammatory response is regulated after an acute injury to the brain.Proinflammatory cytokines of the interleukin-1 (IL-1) family are critical regulators of host responses to infection and orchestrate damaging inflammatory responses that occur during disease (1). One of the main mediators of damaging sterile inflammation is IL-1β, which is implicated in the etiology of many major diseases, including acute cerebrovascular disease (2). Acute cerebrovascular disease presents as a range of conditions, including devastating injuries such as subarachnoid hemorrhage (SAH) and ischemic stroke, which account for up to 10% of mortality worldwide and are the leading cause of morbidity (2). Treatments for acute stroke are limited to thrombolysis for up to 10% of all strokes, antiplatelet agents, and stroke unit care. Thus, treatment of acute cerebrovascular disease remains an area of unmet clinical need. Understanding the mechanisms regulating production of IL-1β during ischemic brain injury may lead to the identification of new therapeutic targets.IL-1β is produced by many cells, most commonly those of macrophage lineage, as a pro–IL-1β precursor. Pro–IL-1β is expressed in response to pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) that bind to pattern recognition receptors (PRRs) to up-regulate proinflammatory gene expression (3, 4). PAMPs are motifs carried by pathogens, such as bacterial endotoxin (or LPS), and DAMPs are commonly endogenous molecules released by necrosis. Pro–IL-1β is inactive and remains intracellular until a further PAMP or DAMP stimulation activates cytosolic PRRs, often of the nucleotide-binding domain and leucine-rich repeat containing receptor (NLR) family, to form large multiprotein complexes called inflammasomes (5). These complexes consist of the PRR, procaspase-1, and, depending upon the PRR, an adaptor protein called ASC, that interact via CARD and pyrin homology-binding domains (5). When the PRR senses PAMPs or DAMPs, it recruits ASC, which in turn recruits caspase-1, causing its activation. Caspase-1 then processes pro–IL-1β to a mature form that is rapidly secreted from the cell (5). The activation of caspase-1 can also cause cell death (6).A number of inflammasome-forming PRRs have been identified, including NLR family, pyrin domain containing 1 (NLRP1); NLRP3; NLRP6; NLRP7; NLRP12; NLR family, CARD domain containing 4 (NLRC4); AIM 2 (absent in melanoma 2); IFI16; and RIG-I (5). Of these inflammasomes identified to date, the best characterized, and most strongly associated with sterile inflammation, is formed by NLRP3 (7). Indeed, there are now several studies suggesting that NLRP3 inflammasomes contribute to ischemic brain injury (8, 9). However, the picture is more complicated. NLRP1 inflammasomes have been implicated in several models of brain injury (6, 10, 11), as have AIM2 inflammasomes, which are suggested to mediate pyroptotic neuronal cell death (12). There is also evidence supporting a role for caspase-1 in brain injury (13), with a selective caspase-1 inhibitor, VRT-018858, a nonpeptide, active metabolite of the prodrug pralnacasan, showing marked protection in a rat model of stroke (14). However, data for the related caspase-1 inhibitor VRT-043198 suggest that it is also an effective inhibitor of caspase-4 (15), a human ortholog of caspase-11. Caspase-11 is also implicated in ischemic brain injury (16, 17), and given that we now also know that the original caspase-1−/− mouse is also deficient in caspase-11 (18), it is clear that caspase-11 could have a role in ischemic brain injury. Our aim here was to elucidate which inflammasomes contribute to ischemic brain injury, using mice in which specific inflammasome components are deleted (−/−).  相似文献   

16.
A series of discrete decanuclear gold(I) μ3-sulfido complexes with alkyl chains of various lengths on the aminodiphosphine ligands, [Au10{Ph2PN(CnH2n+1)PPh2}43-S)4](ClO4)2, has been synthesized and characterized. These complexes have been shown to form supramolecular nanoaggregate assemblies upon solvent modulation. The photoluminescence (PL) colors of the nanoaggregates can be switched from green to yellow to red by varying the solvent systems from which they are formed. The PL color variation was investigated and correlated with the nanostructured morphological transformation from the spherical shape to the cube as observed by transmission electron microscopy and scanning electron microscopy. Such variations in PL colors have not been observed in their analogous complexes with short alkyl chains, suggesting that the long alkyl chains would play a key role in governing the supramolecular nanoaggregate assembly and the emission properties of the decanuclear gold(I) sulfido complexes. The long hydrophobic alkyl chains are believed to induce the formation of supramolecular nanoaggregate assemblies with different morphologies and packing densities under different solvent systems, leading to a change in the extent of Au(I)–Au(I) interactions, rigidity, and emission properties.Gold(I) complexes are one of the fascinating classes of complexes that reveal photophysical properties that are highly sensitive to the nuclearity of the metal centers and the metal–metal distances (159). In a certain sense, they bear an analogy or resemblance to the interesting classes of metal nanoparticles (NPs) (6069) and quantum dots (QDs) (7076) in that the properties of the nanostructured materials also show a strong dependence on their sizes and shapes. Interestingly, while the optical and spectroscopic properties of metal NPs and QDs show a strong dependence on the interparticle distances, those of polynuclear gold(I) complexes are known to mainly depend on the nuclearity and the internuclear separations of gold(I) centers within the individual molecular complexes or clusters, with influence of the intermolecular interactions between discrete polynuclear molecular complexes relatively less explored (3438), and those of polynuclear gold(I) clusters not reported. Moreover, while studies on polynuclear gold(I) complexes or clusters are known (3454), less is explored of their hierarchical assembly and nanostructures as well as the influence of intercluster aggregation on the optical properties (3438). Among the gold(I) complexes, polynuclear gold(I) chalcogenido complexes represent an important and interesting class (4451). While directed supramolecular assembly of discrete Au12 (52), Au16 (53), Au18 (51), and Au36 (54) metallomacrocycles as well as trinuclear gold(I) columnar stacks (3438) have been reported, there have been no corresponding studies on the supramolecular hierarchical assembly of polynuclear gold(I) chalcogenido clusters.Based on our interests and experience in the study of gold(I) chalcogenido clusters (4446, 51), it is believed that nanoaggegrates with interesting luminescence properties and morphology could be prepared by the judicious design of the gold(I) chalcogenido clusters. As demonstrated by our previous studies on the aggregation behavior of square-planar platinum(II) complexes (7780) where an enhancement of the solubility of the metal complexes via introduction of solubilizing groups on the ligands and the fine control between solvophobicity and solvophilicity of the complexes would have a crucial influence on the factors governing supramolecular assembly and the formation of aggregates (80), introduction of long alkyl chains as solubilizing groups in the gold(I) sulfido clusters may serve as an effective way to enhance the solubility of the gold(I) clusters for the construction of supramolecular assemblies of novel luminescent nanoaggegrates.Herein, we report the preparation and tunable spectroscopic properties of a series of decanuclear gold(I) μ3-sulfido complexes with alkyl chains of different lengths on the aminophosphine ligands, [Au10{Ph2PN(CnH2n+1)PPh2}43-S)4](ClO4)2 [n = 8 (1), 12 (2), 14 (3), 18 (4)] and their supramolecular assembly to form nanoaggregates. The emission colors of the nanoaggregates of 2−4 can be switched from green to yellow to red by varying the solvent systems from which they are formed. These results have been compared with their short alkyl chain-containing counterparts, 1 and a related [Au10{Ph2PN(C3H7)PPh2}43-S)4](ClO4)2 (45). The present work demonstrates that polynuclear gold(I) chalcogenides, with the introduction of appropriate functional groups, can serve as building blocks for the construction of novel hierarchical nanostructured materials with environment-responsive properties, and it represents a rare example in which nanoaggregates have been assembled with the use of discrete molecular metal clusters as building blocks.  相似文献   

17.
Cognition presents evolutionary research with one of its greatest challenges. Cognitive evolution has been explained at the proximate level by shifts in absolute and relative brain volume and at the ultimate level by differences in social and dietary complexity. However, no study has integrated the experimental and phylogenetic approach at the scale required to rigorously test these explanations. Instead, previous research has largely relied on various measures of brain size as proxies for cognitive abilities. We experimentally evaluated these major evolutionary explanations by quantitatively comparing the cognitive performance of 567 individuals representing 36 species on two problem-solving tasks measuring self-control. Phylogenetic analysis revealed that absolute brain volume best predicted performance across species and accounted for considerably more variance than brain volume controlling for body mass. This result corroborates recent advances in evolutionary neurobiology and illustrates the cognitive consequences of cortical reorganization through increases in brain volume. Within primates, dietary breadth but not social group size was a strong predictor of species differences in self-control. Our results implicate robust evolutionary relationships between dietary breadth, absolute brain volume, and self-control. These findings provide a significant first step toward quantifying the primate cognitive phenome and explaining the process of cognitive evolution.Since Darwin, understanding the evolution of cognition has been widely regarded as one of the greatest challenges for evolutionary research (1). Although researchers have identified surprising cognitive flexibility in a range of species (240) and potentially derived features of human psychology (4161), we know much less about the major forces shaping cognitive evolution (6271). With the notable exception of Bitterman’s landmark studies conducted several decades ago (63, 7274), most research comparing cognition across species has been limited to small taxonomic samples (70, 75). With limited comparable experimental data on how cognition varies across species, previous research has largely relied on proxies for cognition (e.g., brain size) or metaanalyses when testing hypotheses about cognitive evolution (7692). The lack of cognitive data collected with similar methods across large samples of species precludes meaningful species comparisons that can reveal the major forces shaping cognitive evolution across species, including humans (48, 70, 89, 9398).To address these challenges we measured cognitive skills for self-control in 36 species of mammals and birds (Fig. 1 and Tables S1–S4) tested using the same experimental procedures, and evaluated the leading hypotheses for the neuroanatomical underpinnings and ecological drivers of variance in animal cognition. At the proximate level, both absolute (77, 99107) and relative brain size (108112) have been proposed as mechanisms supporting cognitive evolution. Evolutionary increases in brain size (both absolute and relative) and cortical reorganization are hallmarks of the human lineage and are believed to index commensurate changes in cognitive abilities (52, 105, 113115). Further, given the high metabolic costs of brain tissue (116121) and remarkable variance in brain size across species (108, 122), it is expected that the energetic costs of large brains are offset by the advantages of improved cognition. The cortical reorganization hypothesis suggests that selection for absolutely larger brains—and concomitant cortical reorganization—was the predominant mechanism supporting cognitive evolution (77, 91, 100106, 120). In contrast, the encephalization hypothesis argues that an increase in brain volume relative to body size was of primary importance (108, 110, 111, 123). Both of these hypotheses have received support through analyses aggregating data from published studies of primate cognition and reports of “intelligent” behavior in nature—both of which correlate with measures of brain size (76, 77, 84, 92, 110, 124).Open in a separate windowFig. 1.A phylogeny of the species included in this study. Branch lengths are proportional to time except where long branches have been truncated by parallel diagonal lines (split between mammals and birds ∼292 Mya).With respect to selective pressures, both social and dietary complexities have been proposed as ultimate causes of cognitive evolution. The social intelligence hypothesis proposes that increased social complexity (frequently indexed by social group size) was the major selective pressure in primate cognitive evolution (6, 44, 48, 50, 87, 115, 120, 125141). This hypothesis is supported by studies showing a positive correlation between a species’ typical group size and the neocortex ratio (80, 81, 8587, 129, 142145), cognitive differences between closely related species with different group sizes (130, 137, 146, 147), and evidence for cognitive convergence between highly social species (26, 31, 148150). The foraging hypothesis posits that dietary complexity, indexed by field reports of dietary breadth and reliance on fruit (a spatiotemporally distributed resource), was the primary driver of primate cognitive evolution (151154). This hypothesis is supported by studies linking diet quality and brain size in primates (79, 81, 86, 142, 155), and experimental studies documenting species differences in cognition that relate to feeding ecology (94, 156166).Although each of these hypotheses has received empirical support, a comparison of the relative contributions of the different proximate and ultimate explanations requires (i) a cognitive dataset covering a large number of species tested using comparable experimental procedures; (ii) cognitive tasks that allow valid measurement across a range of species with differing morphology, perception, and temperament; (iii) a representative sample within each species to obtain accurate estimates of species-typical cognition; (iv) phylogenetic comparative methods appropriate for testing evolutionary hypotheses; and (v) unprecedented collaboration to collect these data from populations of animals around the world (70).Here, we present, to our knowledge, the first large-scale collaborative dataset and comparative analysis of this kind, focusing on the evolution of self-control. We chose to measure self-control—the ability to inhibit a prepotent but ultimately counterproductive behavior—because it is a crucial and well-studied component of executive function and is involved in diverse decision-making processes (167169). For example, animals require self-control when avoiding feeding or mating in view of a higher-ranking individual, sharing food with kin, or searching for food in a new area rather than a previously rewarding foraging site. In humans, self-control has been linked to health, economic, social, and academic achievement, and is known to be heritable (170172). In song sparrows, a study using one of the tasks reported here found a correlation between self-control and song repertoire size, a predictor of fitness in this species (173). In primates, performance on a series of nonsocial self-control control tasks was related to variability in social systems (174), illustrating the potential link between these skills and socioecology. Thus, tasks that quantify self-control are ideal for comparison across taxa given its robust behavioral correlates, heritable basis, and potential impact on reproductive success.In this study we tested subjects on two previously implemented self-control tasks. In the A-not-B task (27 species, n = 344), subjects were first familiarized with finding food in one location (container A) for three consecutive trials. In the test trial, subjects initially saw the food hidden in the same location (container A), but then moved to a new location (container B) before they were allowed to search (Movie S1). In the cylinder task (32 species, n = 439), subjects were first familiarized with finding a piece of food hidden inside an opaque cylinder. In the following 10 test trials, a transparent cylinder was substituted for the opaque cylinder. To successfully retrieve the food, subjects needed to inhibit the impulse to reach for the food directly (bumping into the cylinder) in favor of the detour response they had used during the familiarization phase (Movie S2).Thus, the test trials in both tasks required subjects to inhibit a prepotent motor response (searching in the previously rewarded location or reaching directly for the visible food), but the nature of the correct response varied between tasks. Specifically, in the A-not-B task subjects were required to inhibit the response that was previously successful (searching in location A) whereas in the cylinder task subjects were required to perform the same response as in familiarization trials (detour response), but in the context of novel task demands (visible food directly in front of the subject).  相似文献   

18.
19.
Kinesin-1 is a dimeric motor protein, central to intracellular transport, that steps hand-over-hand toward the microtubule (MT) plus-end, hydrolyzing one ATP molecule per step. Its remarkable processivity is critical for ferrying cargo within the cell: over 100 successive steps are taken, on average, before dissociation from the MT. Despite considerable work, it is not understood which features coordinate, or “gate,” the mechanochemical cycles of the two motor heads. Here, we show that kinesin dissociation occurs subsequent to, or concomitant with, phosphate (Pi) release following ATP hydrolysis. In optical trapping experiments, we found that increasing the steady-state population of the posthydrolysis ADP·Pi state (by adding free Pi) nearly doubled the kinesin run length, whereas reducing either the ATP binding rate or hydrolysis rate had no effect. The data suggest that, during processive movement, tethered-head binding occurs subsequent to hydrolysis, rather than immediately after ATP binding, as commonly suggested. The structural change driving motility, thought to be neck linker docking, is therefore completed only upon hydrolysis, and not ATP binding. Our results offer additional insights into gating mechanisms and suggest revisions to prevailing models of the kinesin reaction cycle.Since its discovery nearly 30 years ago (1), kinesin-1—the founding member of the kinesin protein superfamily—has emerged as an important model system for studying biological motors (2, 3). During “hand-over-hand” stepping, kinesin dimers alternate between a two–heads-bound (2-HB) state, with both heads attached to the microtubule (MT), and a one–head-bound (1-HB) state, where a single head, termed the tethered head, remains free of the MT (4, 5). The catalytic cycles of the two heads are maintained out of phase by a series of gating mechanisms, thereby enabling the dimer to complete, on average, over 100 steps before dissociating from the MT (68). A key structural element for this coordination is the neck linker (NL), a ∼14-aa segment that connects each catalytic head to a common stalk (9). In the 1-HB state, nucleotide binding is thought to induce a structural reconfiguration of the NL, immobilizing it against the MT-bound catalytic domain (2, 3, 1017). This transition, called “NL docking,” is believed to promote unidirectional motility by biasing the position of the tethered head toward the next MT binding site (2, 3, 1017). The completion of an 8.2-nm step (18) entails the binding of this tethered head to the MT, ATP hydrolysis, and detachment of the trailing head, thereby returning the motor to the ATP-waiting state (2, 3, 1017). Prevailing models of the kinesin mechanochemical cycle (2, 3, 10, 14, 15, 17), which invoke NL docking upon ATP binding, explain the highly directional nature of kinesin motility and offer a compelling outline of the sequence of events following ATP binding. Nevertheless, these abstractions do not speak directly to the branching transitions that determine whether kinesin dissociates from the MT (off-pathway) or continues its processive reaction cycle (on-pathway). The distance moved by an individual motor before dissociating—the run length—is limited by unbinding from the MT. The propensity for a dimer to unbind involves a competition among multiple, force-dependent transitions in the two heads, which are not readily characterized by traditional structural or bulk biochemical approaches. Here, we implemented high-resolution single-molecule optical trapping techniques to determine transitions in the kinesin cycle that govern processivity.  相似文献   

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

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