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 171 毫秒
1.
2.
Infection of laboratory mice with murine noroviruses (MNV) is widely prevalent. MNV alters various mouse models of disease, including the Helicobacter bilis-induced mouse model of inflammatory bowel disease (IBD) in Mdr1a−/− mice. To further characterize the effect of MNV on IBD, we used mice deficient in the immunoregulatory cytokine IL10 (Il10−/− mice). In vitro infection of Il10−/− bone marrow-derived macrophages (BMDM) with MNV4 cocultured with H. bilis antigens increased the gene expression of the proinflammatory cytokines IL1β, IL6, and TNFα as compared with that of BMDM cultured with H. bilis antigens only. Therefore, to test the hypothesis that MNV4 infection increases inflammation and alters disease phenotype in H. bilis-infected Il10−/− mice, we compared the amount and extent of inflammation in Il10−/− mice coinfected with H. bilis and MNV4 with those of mice singly infected with H. bilis. IBD scores, incidence of IBD, or frequency of severe IBD did not differ between mice coinfected with H. bilis and MNV4 and those singly infected with H. bilis. Mice infected with MNV4 only had no appreciable IBD, comparable to uninfected mice. Our findings suggest that, unlike in Mdr1a−/− mice, the presence of MNV4 in Il10−/− mouse colonies is unlikely to affect the IBD phenotype in a Helicobacter-induced model. However, because MNV4 altered cytokine expression in vitro, our results highlight the importance of determining the potential influence of MNV on mouse models of inflammatory disease, given that MNV has a tropism for macrophages and dendritic cells and that infection is widely prevalent.Abbreviations: BMDM, bone marrow-derived macrophages; IBD, inflammatory bowel disease; MLN, mesenteric lymph node; MNV, murine norovirusInflammatory bowel disease (IBD), which includes both ulcerative colitis and Crohn disease, is a chronic and relapsing inflammatory disorder of the gastrointestinal tract. In addition, patients with IBD may be at increased risk of developing colorectal cancer.15,46 Although the exact mechanisms of disease are still not understood fully, the pathogenesis of disease is likely multifactorial, with components of the innate and adaptive immune systems, host genetics, and environmental factors (for example, the commensal gut microflora) all playing a role.4,37,55Animal models of IBD have been used to advance our knowledge and understanding of IBD pathogenesis and treatment.16,20,37,38,52 One such model that has been widely used to elucidate the mechanisms of IBD is the interleukin10–deficient (Il10−/−) mouse.3,5,6,20,21,29,33,57 The antiinflammatory cytokine IL10 modulates both innate and adaptive immune responses.41 Produced mainly by dendritic cells, monocytes, macrophages, and T regulatory cells, IL10 exerts its immunomodulatory effects by various mechanisms including decreasing secretion of proinflammatory cytokines (for example, interferon γ, IL1, IL2, IL6, IL12 and TNFα) and downregulating important components of innate immune responses and T-cell activation (for example, MHC class II, costimulatory molecules, and nitric oxide production) in antigen presenting cells.14,41 As a consequence, Il10−/− mice, which lack the suppressive effects of IL10, develop IBD in response to their commensal gut microflora or to certain microbial triggers such as Helicobacter infections.5,6,11,21,29,52,57Antigen-presenting cells such as macrophages and dendritic cells play key roles in the inflammatory responses in IBD.32,47,50 In 2003, a newly discovered murine norovirus (MNV) in laboratory mice was shown to infect macrophages and dendritic cells.27,53 Subsequent studies indicated widespread MNV infection in laboratory mice used for biomedical research, with a serologic prevalence as high as 32%.25,43 Members of the genus Norovirus are regarded as gastrointestinal pathogens in humans and animals, eliciting both innate and adaptive immune responses.19 Therefore, in light of the cellular (macrophages and dendritic cells) and tissue (gastrointestinal) tropisms of MNV as well as the high prevalence of MNV infection in laboratory mice, we hypothesized that MNV infection could be a potential confounder in mouse models of inflammatory diseases including IBD. In support of this idea, our laboratory recently reported that MNV infection in Mdr1a−/− mice (FVB.129P2-Abcb1atm1Bor) accelerated weight loss and exacerbated IBD progression initiated by H. bilis infection.31 This effect potentially was mediated in part through modulating dendritic cell and cytokine responses. In addition, others have reported gastrointestinal abnormalities as a result of MNV infection in some strains of mice,7,26,36 whereas others have described the importance of both innate and adaptive immune responses during MNV infection.8,9,10,28,34,36,48 Collectively, these data indicate that MNV could alter inflammatory responses in laboratory mice.Here we extended our studies of MNV beyond Mdr1a−/− mice to Il10−/− mice, another common animal model of IBD, to further examine the potential effect of MNV on IBD research. Disease was initiated in Il10−/− mice with H. bilis, and we determined whether coinfection with MNV altered disease development, incidence, and severity and the production of cytokines. We demonstrated that although MNV stimulates a Th1 skewing of cytokines in Il10−/− bone marrow-derived macrophages (BMDM) in vitro, MNV does not alter the development, incidence, or severity of disease in vivo. Therefore, although MNV may not affect disease in Il10−/− mouse models, the virus may influence in vitro cytokine phenotypes and thus complicate interpretation of such data. To our knowledge, this report is the first to describe the evaluation of MNV infection in the Helicobacter-induced Il10−/− mouse model of IBD.  相似文献   

3.
4.
5.
6.
7.
Caspase-2 has been implicated in various cellular functions, including cell death by apoptosis, oxidative stress response, maintenance of genomic stability and tumor suppression. The loss of the caspase-2 gene (Casp2) enhances oncogene-mediated tumorigenesis induced by E1A/Ras in athymic nude mice, and also in the -Myc lymphoma and MMTV/c-neu mammary tumor mouse models. To further investigate the function of caspase-2 in oncogene-mediated tumorigenesis, we extended our studies in the TH-MYCN transgenic mouse model of neuroblastoma. Surprisingly, we found that loss of caspase-2 delayed tumorigenesis in the TH-MYCN neuroblastoma model. In addition, tumors from TH-MYCN/Casp2−/− mice were predominantly thoracic paraspinal tumors and were less vascularized compared with tumors from their TH-MYCN/Casp2+/+ counterparts. We did not detect any differences in the expression of neuroblastoma-associated genes in TH-MYCN/Casp2−/− tumors, or in the activation of Ras/MAPK signaling pathway that is involved in neuroblastoma progression. Analysis of expression array data from human neuroblastoma samples showed a correlation between low caspase-2 levels and increased survival. However, caspase-2 levels correlated with clinical outcome only in the subset of MYCN-non-amplified human neuroblastoma. These observations indicate that caspase-2 is not a suppressor in MYCN-induced neuroblastoma and suggest a tissue and context-specific role for caspase-2 in tumorigenesis.The caspase family of cysteine proteases are highly conserved regulators of cell death by apoptosis.1 In addition to their pro-apoptotic function, many caspases also have non-apoptotic roles in other physiological processes, such as inflammation, necrosis and tumor suppression.2, 3, 4 The most highly conserved caspase, caspase-2, has recently been demonstrated to function in the cellular stress response, protection against ageing, maintenance of genome stability and in tumor suppression.2, 5, 6, 7, 8The tumor suppressor function of caspase-2 was first demonstrated using E1A/Ras-transformed caspase-2-deficient mouse embryonic fibroblasts (MEFs), which showed an increased tumorigenic potential in athymic nude mice.7 Further supporting evidence came from experiments demonstrating that caspase-2 deficiency enhances B-cell lymphoma development in Eμ-Myc transgenic mice7 and mammary carcinomas in MMTV/c-neu mice,9 suggesting that caspase-2 prevents oncogene-induced lymphomas and epithelial tumors. Importantly, tumor suppression by caspase-2 is also evident in the non-oncogene-driven Atm−/− thymoma mouse model.10Given its role in apoptosis, the tumor suppression function of caspase-2 was thought to be associated with this role, via the elimination of mutagenic or potentially tumorigenic cells. Recent studies have now indicated that the role of caspase-2 may extend beyond apoptosis and that its tumor suppression function may, in part, be mediated by maintaining genomic stability and/or the oxidative stress response. Caspase-2-deficient MEFs and tumor cells from Eμ-Myc/Casp2−/−, MMTV/c-neu/Casp2−/− and Atm−/−;Casp2−/− mice all display aberrant proliferation, and increased genomic instability6, 9, 10 and indicate that caspase-2 is important for the maintenance of genome stability. Importantly, the role of caspase-2 in maintaining genomic stability in primary cells appears to be required for its tumor suppressor function.10Genomic instability is a hallmark of cancer11 and the overexpression of Myc family oncoproteins is commonly associated with genomic instability and a wide spectrum of human cancers.12, 13, 14 Interestingly, a common feature of the oncogene-induced tumor models used in the study of caspase-2 tumor suppressor function is the overexpression of c-Myc15 or aberrant c-Myc signaling.16, 17, 18 Given the role of Myc proteins as key mediators of genomic instability as well as cell proliferation, cell growth and DNA damage, we were interested in further assessing whether caspase-2 can promote tumor suppression in other MYC-dependent mouse tumor models. We used the MYCN mouse model of neuroblastoma (TH-MYCN mouse), in which MYCN is constitutively expressed under the control of the rat tyrosine hydroxylase (TH) promoter leading to neural crest cell-specific expression and early-onset neuroblastoma.19 Amplification of MYCN occurs in ∼20% of human neuroblastomas and high MYCN protein levels are strongly associated with tumor progression and poor clinical outcome.20, 21 Thus, the TH-MYCN transgenic mouse model recapitulates many clinical features of aggressive neuroblastomas in humans and provides a powerful model of preclinical neuroblastoma.19, 22MYCN-mediated neuroblastoma onset and progression is commonly associated with additional genetic events, including the expression of the key genes including Odc1, Mrp1, SirT1 and Ras.23, 24, 25 A recent study has found that caspase-8 is in fact a potent suppressor of neuroblastoma, with the loss of caspase-8 expression occurring in ∼70% of neuroblastoma patients.26, 27 Interestingly, the loss of caspase-8 also promotes bone marrow metastasis in the TH-MYCN neuroblastoma mouse model.26, 27 The role of other caspases in neuroblastoma has not previously been examined, and given the function of caspase-2 in tumor suppression, provided additional relevance in assessing its role in this model.This study shows that caspase-2 is not able to suppress neuroblastoma development in TH-MYCN mice. In contrast to a role for caspase-2 as a tumor suppressor, our findings demonstrate that loss of caspase-2 somewhat delays neuroblastoma onset in mice. Interestingly, expression array data from human neuroblastoma show a strong correlation between low caspase-2 levels and improved outcome. Our data demonstrate that the tumor suppressor function of caspase-2 is not specific to Myc-mediated oncogenesis and that its role is likely to be tissue- and/or context-specific.  相似文献   

8.
M Shen  L Wang  B Wang  T Wang  G Yang  L Shen  T Wang  X Guo  Y Liu  Y Xia  L Jia  X Wang 《Cell death & disease》2014,5(11):e1528
Endoplasmic reticulum (ER) stress occurring in stringent conditions is critically involved in cardiomyocytes apoptosis and cardiac contractile dysfunction (CCD). However, the molecular machinery that mediates cardiac ER stress and subsequent cell death remains to be fully deciphered, which will hopefully provide novel therapeutic targets for these disorders. Here, we establish tunicamycin-induced model of cardiomyocyte ER stress, which effectively mimicks pathological stimuli to trigger CCD. Tunicamycin activates volume-sensitive outward rectifying Cl currents. Blockade of the volume-sensitive outwardly rectifying (VSOR) Cl channel by 4,4''-diisothiocya-natostilbene-2,2''-disulfonic acid (DIDS), a non-selective Cl channel blocker, and 4-(2-butyl-6,7-dichlor-2-cyclopentyl-indan-1-on-5-yl) oxybutyric acid (DCPIB), a selective VSOR Cl channel blocker, improves cardiac contractility, which correlates with suppressed ER stress through inhibiting the canonical GRP78/eIF2α/ATF4 and XBP1 pathways, and promotes survival of cardiomyocytes by inverting tunicamycin-induced decrease of Wnt through the CHOP pathway. VSOR activation of tunicamycin-treated cardiomyocytes is attributed to increased intracellular levels of reactive oxygen species (ROS). Our study demonstrates a pivotal role of ROS/VSOR in mediating ER stress and functional impairment of cardiomyocytes via the CHOP-Wnt pathway, and suggests the therapeutic values of VSOR Cl channel blockers against ER stress-associated cardiac anomalies.The endoplasmic reticulum (ER) is characterized as an organelle that participates in the folding of membrane and secretory proteins.1,2 Efficient functioning of the endoplasmic reticulum is important for cell function and survival. Perturbations of ER homeostasis by energy deprivation and glucose,3 viral infections4 and accumulation of misfolded and/or unfolded proteins2 interfere with ER function, leading to a state of ER stress.5, 6, 7 A cohort of chemicals, for example, tunicamycin and thapsigargin, also trigger ER stress.8, 9, 10 Thapsigargin disrupts the calcium storage of ER by blocking calcium reuptake into the ER lumen, thus by depleting calcium from the organelle.11 In particular, tunicamycin is a highly specific ER stress inducer by inhibiting N-linked glycosylation of protein, representing a well-documented method to artificially elicit unfolded protein response.8 In response to ER stress, ER chaperones such as glucose-regulated protein 78 kDa (GRP78) and glucose-regulated protein 94 kDa (GRP94) are upregulated to facilitate the recovery of unfolded or misfolded proteins.12 ER stress may act as a defense mechanism against external insults; however, prolonged and/or severe ER stress may ultimately trigger apoptosis.8 The C/EBP homologous protein (CHOP) has been defined as a pivotal mediator of cell death signaling in ER stress.13, 14 Accumulating evidence has demonstrated that ER stress-induced cell death is an essential step in the pathogenesis of a wide variety of cardiovascular diseases such as ischemia reperfusion heart diseases,15 atherosclerosis,5, 16, 17, 18 myocardial infarction,19 hypertension20, 21 and heart failure.8, 22, 23 Inhibiting ER stress has great therapeutic values for cardiac anomalies. However, the precise mechanism involved in ER stress-induced cardiovascular diseases has not been well identified, which impedes the translation of our understanding of ER stress-induced cardiovascular anomalies into effective therapeutic strategies. Apoptosis induction requires persistent cell shrinkage, named apoptotic volume decrease (AVD).24, 25, 26, 27 It is an early prerequisite for the activation of caspases.24 In various types of cells including cardiomyocytes, AVD process is accomplished by the activation of volume-sensitive outwardly rectifying (VSOR) Cl channel and is concomitant with the egress of water from the cells undergoing mitochondrion-initiated or death receptor-induced apoptosis.25, 28, 29, 30 Although inhibition of VSOR Cl channel by DIDS (4,4''-diisothiocyanatostilbene-2,2''-disulphonic acid) and DCPIB (4-(2-butyl-6,7- dichlor-2-cyclopentyl-indan-1-on-5-yl) oxybutyric acid) blocked AVD and rescued cardiomyocytes from mitochondrial and death receptor pathway-induced apoptosis,31, 32 it remains largely unknown concerning the role of VSOR Cl channel and how it is regulated in ER stress-induced apoptotic cardiomyocyte death.Emerging evidence indicates that Wnt signal pathways are found to be anti-apoptotic in the cardiovascular diseases,33, 34, 35 regulating crucial aspects of cardiovascular biology. However, up to now, its activity in ER stress-induced apoptosis and in the process of AVD in cardiomyocytes remains elusive.In the present study, we probed the role of VSOR Cl channel in ER stress-induced apoptosis of cardiomyocytes, which intimately correlates with cardiac contractile dysfunction (CCD). We hypothesized that VSOR Cl channel controls the process of AVD occurring concomitantly with ER stress-induced apoptosis of cardiomyocytes. To test this hypothesis, we investigated VSOR Cl currents in cardiomyocytes treated with the ER stress inducer tunicamycin. The pathophysiological role of VSOR Cl channel and the potential signaling mechanisms in the development of ER stress-induced apoptosis in CCD were also dissected.  相似文献   

9.
Neurodegeneration is a serious issue of neurodegenerative diseases including epilepsy. Downregulation of the chloride transporter KCC2 in the epileptic tissue may not only affect regulation of the polarity of GABAergic synaptic transmission but also neuronal survival. Here, we addressed the mechanisms of KCC2-dependent neuroprotection by assessing truncated and mutated KCC2 variants in different neurotoxicity models. The results identify a threonine- and tyrosine-phosphorylation-resistant KCC2 variant with increased chloride transport activity, but they also identify the KCC2 N-terminal domain (NTD) as the relevant minimal KCC2 protein domain that is sufficient for neuroprotection. As ectopic expression of the KCC2-NTD works independently of full-length KCC2-dependent regulation of Cl transport or structural KCC2 C-terminus-dependent regulation of synaptogenesis, our study may pave the way for a selective neuroprotective therapeutic strategy that will be applicable to a wide range of neurodegenerative diseases.Neurodegeneration restricts neuron numbers during development but can become a serious issue in disease conditions such as temporal lobe epilepsy (TLE).1 GABA-activated Cl channels contribute to activity-dependent refinement of neural networks by triggering the so-called giant depolarizing potentials providing developing neurons with a sense of activity essential for neuronal survival and co-regulation of excitatory glutamatergic and (inhibitory) GABAergic synapses.2 By regulating transmembrane Cl gradients KCC2 plays a vital role in development and disease.3 In addition, KCC2 plays a protein structural role in spine formation through its C-terminal protein domain (CTD).4, 5 Hence, regulation of KCC2 expression and function is relevant for development and disease-specific plasticity of neural networks.6, 7, 8, 9GlyR α3K RNA editing leads to proline-to-leucine substitution (P185L) in the ligand-binding domain and generates gain-of-function neurotransmitter receptors.10, 11, 12, 13 GlyR RNA editing is upregulated in the hippocampus of patients with TLE and leads to GlyR α3K185L-dependent tonic inhibition of neuronal excitability associated with neurodegeneration.14 KCC2 expression promotes neuroprotection14, 15 but whether this involves regulation of transmembrane Cl gradient or protein structural role is a matter of debate.14, 15Here, we assessed neuroprotection through several KCC2 variants in two different models of neurodegeneration including chronic neuronal silencing (α3K185L model) and acute neuronal overexcitation (NMDA model).14, 15 The results identify a threonine- and tyrosine-phosphorylation-resistant KCC2 variant with increased Cl transport activity, but they also demonstrate that the N-terminal KCC2 protein domain (NTD) is sufficient for neuroprotection.  相似文献   

10.
11.
Although cellular prion protein (PrPc) has been suggested to have physiological roles in neurogenesis and angiogenesis, the pathophysiological relevance of both processes remain unknown. To elucidate the role of PrPc in post-ischemic brain remodeling, we herein exposed PrPc wild type (WT), PrPc knockout (PrP−/−) and PrPc overexpressing (PrP+/+) mice to focal cerebral ischemia followed by up to 28 days reperfusion. Improved neurological recovery and sustained neuroprotection lasting over the observation period of 4 weeks were observed in ischemic PrP+/+ mice compared with WT mice. This observation was associated with increased neurogenesis and angiogenesis, whereas increased neurological deficits and brain injury were noted in ischemic PrP−/− mice. Proteasome activity and oxidative stress were increased in ischemic brain tissue of PrP−/− mice. Pharmacological proteasome inhibition reversed the exacerbation of brain injury induced by PrP−/−, indicating that proteasome inhibition mediates the neuroprotective effects of PrPc. Notably, reduced proteasome activity and oxidative stress in ischemic brain tissue of PrP+/+ mice were associated with an increased abundance of hypoxia-inducible factor 1α and PACAP-38, which are known stimulants of neural progenitor cell (NPC) migration and trafficking. To elucidate effects of PrPc on intracerebral NPC homing, we intravenously infused GFP+ NPCs in ischemic WT, PrP−/− and PrP+/+ mice, showing that brain accumulation of GFP+ NPCs was greatly reduced in PrP−/− mice, but increased in PrP+/+ animals. Our results suggest that PrPc induces post-ischemic long-term neuroprotection, neurogenesis and angiogenesis in the ischemic brain by inhibiting proteasome activity.Endogenous neurogenesis persists in the adult rodent brain within distinct niches such as the subventricular zone (SVZ) of the lateral ventricles,1, 2, 3, 4 which host astrocyte-like neural stem cells and neural progenitor cells (NPCs). Focal cerebral ischemia stimulates neurogenesis, and NPCs proliferate and migrate towards the site of lesion where they eventually differentiate.5, 6, 7 In light of low differentiation rates and high cell death rates of new-born cells,6, 8, 9 post-stroke neurogenesis is scarce.10Cellular prion protein (PrPc) is a glycoprotein that is attached to cell membranes by means of a glycosylphosphatidylinositol anchor.11 Although PrPc is ubiquitously expressed, it is most abundant within the central nervous system. Conversion into its misfolded isoform PrPsc causes neurodegenerative diseases such as Creutzfeldt-Jacob disease.11, 12 While a large body of studies analyzed the role of PrPsc in the context of transmissible spongiform encephalopathies, little is known about the physiological role of PrPc. Studies performed during both ontogenesis and adulthood suggest that PrPc regulates neuronal proliferation and differentiation, synaptic plasticity and angiogenesis.13, 14, 15, 16, 17, 18 The role of these processes under pathophysiological conditions, however, is largely unknown.Previous reports suggested a role of PrPc in post-ischemic neuroprotection.19, 20, 21, 22, 23, 24 Thus, PrPc was found to be overexpressed in ischemic brain tissue.19, 20, 21, 22, 23, 24 PrPc deficiency aggravated ischemic brain injury, possibly via enhanced ERK-1/2 activation and reduced phosphorylation of Akt, thus ultimately culminating in increased caspase-3 activity,21, 24 whereas PrPc overexpression protected against ischemia.19, 20, 21, 22, 23, 24 Nevertheless, these studies focused on acute injury processes with a maximal observation period of 3 days, leaving the biological role of PrPc in post-stroke neurogenesis and angiogenesis unanswered. To clarify the role of PrPc in the post-acute ischemic brain, we herein exposed PrPc wild type (WT), PrPc knockout (PrP−/−) and PrPc overexpressing (PrP+/+) mice to focal cerebral ischemia induced by intraluminal middle cerebral artery (MCA) occlusion, evaluating effects of PrPc on neurological recovery, ischemic injury, neurogenesis and angiogenesis, as well as the homing and efficacy of exogenously delivered NPCs.  相似文献   

12.
Mycoplasmosis is a frequent causative microbial agent of community-acquired pneumonia and has been linked to exacerbation of chronic obstructive pulmonary disease. The macrophage class A scavenger receptor (SRA) facilitates the clearance of noxious particles, oxidants, and infectious organisms by alveolar macrophages. We examined wildtype and SRA−/− mice, housed in either individually ventilated or static filter-top cages that were cycled with fresh bedding every 14 d, as a model of gene–environment interaction on the outcome of pulmonary Mycoplasma pulmonis infection. Intracage NH3 gas measurements were recorded daily prior to infection. Mice were intranasally infected with 1 × 107 cfu M. pulmonis UAB CT and evaluated at 3, 7, and 14 d after inoculation. Wildtype mice cleared 99.5% of pulmonary M. pulmonis by 3 d after infection but remained chronically infected through the study. SRA−/− mice were chronically infected with 40-fold higher mycoplasma numbers than were wildtype mice. M. pulmonis caused a chronic mixed inflammatory response that was accompanied with high levels of IL1β, KC, MCP1, and TNFα in SRA−/− mice, whereas pulmonary inflammation in WT mice was represented by a monocytosis with elevation of IL1β. Housing had a prominent influence on the severity and persistence of mycoplasmosis in SRA−/− mice. SRA-/- mice housed in static cages had an improved recovery and significant changes in surfactant proteins SPA and SPD compared with baseline levels. These results indicate that SRA is required to prevent chronic mycoplasma infection of the lung. Furthermore, environmental conditions may exacerbate chronic inflammation in M. pulmonis-infected SRA−/− mice.Abbreviations: BAL, bronchoalveolar lavage; COPD, chronic obstructive pulmonary disease; KC, keratinocyte-derived chemokine (CXCL1); MCP1, monocyte chemotactic protein 1; SPA, surfactant protein A (SFTPA1); SPB, surfactant protein B (SFTPB); SPD, surfactant protein D (SFTPD); SRA, class A scavenger receptor (MSR1); WT, wildtypeThere are numerous options for the housing and husbandry of rodents in the laboratory setting. Various available choices in caging, bedding material, and cage-change frequency have the potential to effect physiologic values and thus experimental outcomes.20,108 In many facilities, current practices involve performing cage changes every 1 to 2 wk, with some facilities exploring the possibility of extending these practices to every 4 wk.97 Cage-change frequency practices are established at various institutions after consideration of several variables that affect animal health, welfare, and cost. Ideally, an appropriate sanitation program provides clean and dry bedding, adequate air quality, and clean cage surfaces and accessories.44 When establishing performance standards for a sanitation program that are different from those which are recommended in the Guide for the Care and Use of Animals in Research,44 microenvironmental conditions, including intracage humidity, temperature, animal behavior and appearance, microbiologic loads, and levels of pollutants such as CO2 and NH3, should be evaluated and verified. Although there are currently no established NH3 exposure limits for laboratory animals, the human occupational exposure limit of 25 ppm as an 8-h time-weighted average, established by the National Institute for Occupational Safety and Health, is often referenced as a guideline for animals.95 Multiple factors, such as animal cage density, sex, age, bedding type, reusable compared with disposable caging, static caging compared with IVC, and cage-change frequency, influence intracage and ambient NH3 levels.82,83,97 Only limited information is available that addresses the effect of natural intracage NH3 levels on respiratory function in experimental rodents and whether exposure to high NH3 levels under current standard practices affects the results of respiratory disease research.Ammonia is an alkaline, corrosive, and irritant gas that is very water soluble. It reacts with the moisture of the mucous membranes of the eyes, mouth, and respiratory tract to form ammonium hydroxide in an exothermic reaction, resulting in thermal and chemical burns.68 Clinical symptoms in humans exposed to high levels of NH3 include eye irritation, headaches, and multiple acute and chronic respiratory symptoms, such as irritation of the nose, pharynx, and sinuses, and in severe cases, development of bronchitis and hyper-reactive airway disease.79 Animals are similarly susceptible to NH3-induced pulmonary disease.23,31,48Mice exposed to naturally increasing levels of intracage NH3 can develop lesions in the rostral nasal cavity, with decreasing severity of the lesions moving caudally into the nasopharynx, and no lesions in the lung.97 However, dust is another common environmental pollutant that is often present in animal settings. Dust particles readily absorb NH3, which then serve as a source of NH3 deposition into the lower respiratory tract. Dust particulate can range from large (300 µm), minimally respirable particles to very fine (< 50 µm) particulate matter, which can settle deep within the alveoli.10,102 The mucociliary system of the respiratory tract is the first line of defense against inspired noxious stimuli and pathogens. Exposure of the ciliated respiratory epithelium to the damaging effects of NH3 are known to cause decreased mucociliary beating.56 Disruption of the respiratory mucociliary escalator initiated by NH3 exposure can then promote establishment of chronic infections and inflammation of the airway mucosa.11,87 Therefore, NH3 potentially can cause pathophysiologic changes of the lung in the absence of histopathologic lesions.Our primary goal was to analyze the effect of 2 housing modalities, which result in different intracage NH3 concentrations, on mice that were challenged with a respiratory pathogen. Mycoplasma pulmonis was chosen as a model because it is a well-established model in rodents which causes chronic mycoplasmosis and reproduces the features of M. pneumoniae in humans.22,41 M. pneumoniae infection is a frequent and contagious etiology of community-acquired pneumonia causing tracheobronchitis, sneezing, cough, and inflammation of the respiratory tract.8,12,47,63 Moreover, atypical and difficult-to-detect respiratory pathogens such as Chlamydophila pneumoniae and Mycoplasma pneumoniae that can establish chronic asymptomatic infections may contribute to both the development and exacerbation of COPD26,45,57,58,62,63,66,72,96,103 and asthma.8,51,65 Infection with M. pulmonis in rodents causes rhinitis, otitis media, tracheitis, and pneumonia, which can be exacerbated by housing conditions and genetic background.14,32,85 The mechanism of pathogenicity of mycoplasmas continues to be an area of interest in the research.The innate host factors protecting against pulmonary mycoplasmosis include the secreted surfactant protein opsonins SPA and SPD, surfactant phospholipids, and the molecular pattern-recognition receptor TLR2.15,16,54,74 Therefore, compared with their wildtype (WT) counterparts, SPA-deficient mice infected with either M. pulmonis or M. pneumoniae develop more severe inflammation and have decreased capacity to clear these infections from the lungs.43 In addition, TLR2-deficient mice exhibit decreased clearance and increased inflammation in response to mycoplasma infection.60,104Second, we wanted to study the effects of SRA deficiency in mycoplasmosis. The class A scavenger receptor (SRA) modulates inflammatory responses and mediates the clearance of airborne oxidants, particulates, and respiratory pathogens.3,17,18,49,88,101 Inhibition of SRA expression in alveolar macrophages in an elastase–LPS model of COPD was associated with decreased clearance of Haemophilus influenzae.33 Lack of SRA similarly impaired alveolar macrophage-mediated clearance of Streptococcus pneumoniae,5 environmental particles,6 and ozone-oxidized lipids18 by alveolar macrophages. Absence of SRA also enhanced hyperoxia-induced lung injury49 and exacerbated inflammation in response to Staphylococcus aureus infection.88 SRA appears to have antiinflammatory properties with the capacity to modify macrophage phenotype and suppress polarization toward the M1 alternative macrophage activation state.13 The SRA gene (MSR1) is polymorphic in both mice and humans.19,29,105 Genetic association studies in humans, however, showed that subjects with truncations or point mutations in MSR1 have significantly increased risk for the development of pulmonary diseases such as COPD33,38,71,94 and asthma.5 Our understanding of the immune factors that contribute to mycoplasmosis is far from complete.In the present study, by investigating the role of SRA in mycoplasmosis jointly with the effects of housing, we demonstrated that genetic and environmental factors both serve as critical players in disease progression. We show that SRA-deficient mice are susceptible to chronic colonization with M. pulmonis and development of chronic mycoplasma-induced bronchopneumonia characterized by persistent multicellular inflammation. Furthermore, we show that housing conditions influence the effect of SRA deficiency on the severity of mycoplasmosis. Taken together, these results indicate that lack of SRA function impairs host protection against both infectious and environmental insults.  相似文献   

13.
Tumor necrosis factor α (TNFα) triggers necroptotic cell death through an intracellular signaling complex containing receptor-interacting protein kinase (RIPK) 1 and RIPK3, called the necrosome. RIPK1 phosphorylates RIPK3, which phosphorylates the pseudokinase mixed lineage kinase-domain-like (MLKL)—driving its oligomerization and membrane-disrupting necroptotic activity. Here, we show that TNF receptor-associated factor 2 (TRAF2)—previously implicated in apoptosis suppression—also inhibits necroptotic signaling by TNFα. TRAF2 disruption in mouse fibroblasts augmented TNFα–driven necrosome formation and RIPK3-MLKL association, promoting necroptosis. TRAF2 constitutively associated with MLKL, whereas TNFα reversed this via cylindromatosis-dependent TRAF2 deubiquitination. Ectopic interaction of TRAF2 and MLKL required the C-terminal portion but not the N-terminal, RING, or CIM region of TRAF2. Induced TRAF2 knockout (KO) in adult mice caused rapid lethality, in conjunction with increased hepatic necrosome assembly. By contrast, TRAF2 KO on a RIPK3 KO background caused delayed mortality, in concert with elevated intestinal caspase-8 protein and activity. Combined injection of TNFR1-Fc, Fas-Fc and DR5-Fc decoys prevented death upon TRAF2 KO. However, Fas-Fc and DR5-Fc were ineffective, whereas TNFR1-Fc and interferon α receptor (IFNAR1)-Fc were partially protective against lethality upon combined TRAF2 and RIPK3 KO. These results identify TRAF2 as an important biological suppressor of necroptosis in vitro and in vivo.Apoptotic cell death is mediated by caspases and has distinct morphological features, including membrane blebbing, cell shrinkage and nuclear fragmentation.1, 2, 3, 4 In contrast, necroptotic cell death is caspase-independent and is characterized by loss of membrane integrity, cell swelling and implosion.1, 2, 5 Nevertheless, necroptosis is a highly regulated process, requiring activation of RIPK1 and RIPK3, which form the core necrosome complex.1, 2, 5 Necrosome assembly can be induced via specific death receptors or toll-like receptors, among other modules.6, 7, 8, 9 The activated necrosome engages MLKL by RIPK3-mediated phosphorylation.6, 10, 11 MLKL then oligomerizes and binds to membrane phospholipids, forming pores that cause necroptotic cell death.10, 12, 13, 14, 15 Unchecked necroptosis disrupts embryonic development in mice and contributes to several human diseases.7, 8, 16, 17, 18, 19, 20, 21, 22The apoptotic mediators FADD, caspase-8 and cFLIP suppress necroptosis.19, 20, 21, 23, 24 Elimination of any of these genes in mice causes embryonic lethality, subverted by additional deletion of RIPK3 or MLKL.19, 20, 21, 25 Necroptosis is also regulated at the level of RIPK1. Whereas TNFα engagement of TNFR1 leads to K63-linked ubiquitination of RIPK1 by cellular inhibitor of apoptosis proteins (cIAPs) to promote nuclear factor (NF)-κB activation,26 necroptosis requires suppression or reversal of this modification to allow RIPK1 autophosphorylation and consequent RIPK3 activation.2, 23, 27, 28 CYLD promotes necroptotic signaling by deubiquitinating RIPK1, augmenting its interaction with RIPK3.29 Conversely, caspase-8-mediated CYLD cleavage inhibits necroptosis.24TRAF2 recruits cIAPs to the TNFα-TNFR1 signaling complex, facilitating NF-κB activation.30, 31, 32, 33 TRAF2 also supports K48-linked ubiquitination and proteasomal degradation of death-receptor-activated caspase-8, curbing apoptosis.34 TRAF2 KO mice display embryonic lethality; some survive through birth but have severe developmental and immune deficiencies and die prematurely.35, 36 Conditional TRAF2 KO leads to rapid intestinal inflammation and mortality.37 Furthermore, hepatic TRAF2 depletion augments apoptosis activation via Fas/CD95.34 TRAF2 attenuates necroptosis induction in vitro by the death ligands Apo2L/TRAIL and Fas/CD95L.38 However, it remains unclear whether TRAF2 regulates TNFα-induced necroptosis—and if so—how. Our present findings reveal that TRAF2 inhibits TNFα necroptotic signaling. Furthermore, our results establish TRAF2 as a biologically important necroptosis suppressor in vitro and in vivo and provide initial insight into the mechanisms underlying this function.  相似文献   

14.
15.
Bak and Bax mediate apoptotic cell death by oligomerizing and forming a pore in the mitochondrial outer membrane. Both proteins anchor to the outer membrane via a C-terminal transmembrane domain, although its topology within the apoptotic pore is not known. Cysteine-scanning mutagenesis and hydrophilic labeling confirmed that in healthy mitochondria the Bak α9 segment traverses the outer membrane, with 11 central residues shielded from labeling. After pore formation those residues remained shielded, indicating that α9 does not line a pore. Bak (and Bax) activation allowed linkage of α9 to neighboring α9 segments, identifying an α9:α9 interface in Bak (and Bax) oligomers. Although the linkage pattern along α9 indicated a preferred packing surface, there was no evidence of a dimerization motif. Rather, the interface was invoked in part by Bak conformation change and in part by BH3:groove dimerization. The α9:α9 interaction may constitute a secondary interface in Bak oligomers, as it could link BH3:groove dimers to high-order oligomers. Moreover, as high-order oligomers were generated when α9:α9 linkage in the membrane was combined with α6:α6 linkage on the membrane surface, the α6-α9 region in oligomerized Bak is flexible. These findings provide the first view of Bak carboxy terminus (C terminus) membrane topology within the apoptotic pore.Mitochondrial permeabilization during apoptosis is regulated by the Bcl-2 family of proteins.1, 2, 3 Although the Bcl-2 homology 3 (BH3)-only members such as Bid and Bim trigger apoptosis by binding to other family members, the prosurvival members block apoptosis by sequestering their pro-apoptotic relatives. Two remaining members, Bak and Bax, form the apoptotic pore within the mitochondrial outer membrane (MOM).Bak and Bax are globular proteins comprising nine α-helices.4, 5 They are activated by BH3-only proteins binding to the α2–α5 surface groove,6, 7, 8, 9, 10, 11, 12 or for Bax, to the α1/α6 ‘rear pocket''.13 Binding triggers dissociation of the latch domain (α6–α8) from the core domain (α2–α5), together with exposure of N-terminal epitopes and the BH3 domain.6, 7, 14, 15, 16 The exposed BH3 domain then binds to the hydrophobic groove in another Bak or Bax molecule to generate symmetric homodimers.6, 7, 14, 17, 18 In addition to dimerizing, parts of activated Bak and Bax associate with the lipid bilayer.19 In Bax, the α5 and α6 helices may insert into the MOM,20 although recent studies indicate that they lie in-plane on the membrane surface, with the hydrophobic α5 sandwiched between the membrane and a BH3:groove dimer interface.7, 21, 22, 23 The dimers can be linked via cysteine residues placed in α6,18, 24, 25 and more recently via cysteine residues in either α3 or α5,6, 21 allowing detection of the higher-order oligomers associated with pore formation.26, 27 However, whether these interactions are required for high-order oligomers and pore formation remains unclear.Like most Bcl-2 members, Bak and Bax are targeted to the MOM via a hydrophobic C-terminal region. The C terminus targets Bak to the MOM in healthy cells,28 whereas the Bax C terminus is either exposed29 or sequestered within the hydrophobic groove until apoptotic signals trigger Bax translocation.5, 30, 31 The hydrophobic stretch is important, as substituting polar or charged residues decreased targeting of Bak and Bax.10, 32 Mitochondrial targeting is also controlled by basic residues at the far C termini,32, 33, 34 and by interaction with VDAC235, 36 via the Bak and Bax C termini.37, 38 Retrotranslocation of Bak and Bax was also altered by swapping the C termini.39The membrane topology of the Bak and Bax C termini before and after apoptosis has not been examined directly, due in part to difficulty in reconstituting oligomers of full-length Bak in artificial membranes. Nor is it known whether the C termini contribute to pore formation by promoting oligomerization or disturbing the membrane. To address these questions synthetic peptides based on the Bak and Bax C termini have been studied in model membranes. The peptides adopt a predominantly α-helical secondary structure,40, 41, 42, 43 with orientation affected by lipid composition.42, 44, 45 The peptides could also permeabilize lipid vesicles,41, 43, 46, 47 suggesting that the C termini in full-length Bak and Bax may contribute to pore formation.Here we examined the membrane topology of the C termini within full-length Bak and Bax in the MOM, both before and after apoptotic pore formation. After pore formation the α9 helices of Bak (and of Bax) became juxtaposed but did not line the surface of a pore. The α9:α9 interaction occurred after Bak activation and conformation change, but was promoted by formation of BH3:groove dimers. Combining linkage at more than one interface indicated that the Bak α9:α9 interface can link BH3:groove dimers to high-order oligomers, and moreover, that the α6–α9 region is flexible in oligomerized Bak.  相似文献   

16.
The death receptor, Fas, triggers apoptotic death and is essential for maintaining homeostasis in the peripheral lymphoid organs. RIP1 was originally cloned when searching for Fas-binding proteins and was later shown to associate also with the signaling complex of TNFR1. Although Fas exclusively induces apoptosis, TNFR1 primarily activates the pro-survival/pro-inflammatory NF-κB pathway. Mutations in Fas lead to lymphoproliferative (lpr) diseases, and deletion of TNFR1 results in defective innate immune responses. However, the function of RIP1 in the adult lymphoid system has not been well understood, primarily owing to perinatal lethality in mice lacking the entire RIP1 protein in germ cells. This current study investigated the requirement for RIP1 in the T lineage using viable RIP1 mutant mice containing a conditional and kinase-dead RIP1 allele. Disabling the kinase activity of RIP1 had no obvious impact on the T-cell compartment. However, T-cell-specific deletion of RIP1 led to a severe T-lymphopenic condition, owing to a dramatically reduced mature T-cell pool in the periphery. Interestingly, the immature T-cell compartment in the thymus appeared intact. Further analysis showed that mature RIP1−/− T cells were severely defective in antigen receptor-induced proliferative responses. Moreover, the RIP1−/− T cells displayed greatly increased death and contained elevated caspase activities, an indication of apoptosis. In total, these results revealed a novel, kinase-independent function of RIP1, which is essential for not only promoting TCR-induced proliferative responses but also in blocking apoptosis in mature T cells.The pro-survival signaling pathways provide protection against cell death responses at various stages during T lymphopoiesis as well as maintenance of the mature population.1, 2 Apoptosis is a major programmed cell death pathway, which can be induced through either intrinsic or extrinsic signals.3 Under normal circumstances, the pro-survival and apoptosis signaling pathways are tightly regulated, which ensures generation of diverse T-cell repertoires, while avoiding autoimmunity. For instance, the Bcl-2 and Bcl-XL genes, which inhibit the intrinsic apoptotic pathway, are essential for both T-cell development and peripheral maintenance.4, 5 However, lack of cell death, as in the case of inactivation of Bim, a pro-apoptotic protein of the Bcl-2 family, results in lymphoproliferative and autoimmune diseases.6 The extrinsic pathway of apoptosis is triggered through cell receptors, including Fas/Apo-1 and tumor necrosis factor receptor 1 (TNFR1). Whereas Fas is a professional death receptor, TNFR1 primarily signals the pro-survival pathway by activating NF-κB, which also promotes inflammation.7, 8Receptor-interacting protein (RIP or RIP1) was originally cloned as a potential Fas-interacting protein.9 However, later studies found that lack of RIP1 has no effect on Fas-induced apoptosis.10, 11 Subsequently, RIP1 was also found to associate with the signaling complex of TNFR1.12 It was shown that RIP1 deficiency disrupts NF-κB activation induced by TNFR1 in primary mouse embryonic fibroblast cells or human Jurkat T lymphoma cells.10, 11 Several functional domains of RIP1 have been defined. In particular, RIP1 contains a serine/threonine kinase domain (KD) at the amino-terminus and a death domain (DD) at the carboxy-terminus. The intermediate domain, but not the protein serine/threonine KD of RIP1, is required for the activation of NF-κB.13 The DD of RIP1 interacts with the DD of TNFR1-associated death domain (TRADD) protein, a signaling adaptor, leading to both apoptosis and NF-κB activation.12 Therefore, RIP1 may serve as a scaffold protein in addition to being a protein serine/threonine kinase.The function of the KD of RIP1 remained unknown until the landmark work by Holler et al.,14 implicating a novel function for RIP1 in a caspase-independent cell death process with certain characteristics of necrosis, namely necroptosis. Importantly, mutations targeting the kinase activity of RIP1 abolish necroptotic cell death induced by TNFR1. The in vivo role of RIP1-mediated necroptosis was first revealed by analysis of the embryonic defect displayed by mice lacking the Fas-associated death domain (FADD) protein.15 The FADD adaptor protein relays exclusively apoptotic signals in the pathways triggered by Fas, TNFR1, and TNF-related apoptosis-inducing ligand receptors (TRAIL-Rs or DR4/5).16, 17, 18 Whereas none of the DRs are essential for mouse development, FADD deficiency resulted in midgestation death of mouse embryos.19, 20 Interestingly, when RIP1 is absent, normal embryonic development is restored in FADD−/− mice,15 indicating that FADD−/− embryonic lethality is caused by RIP1-dependent necroptosis.Although normal during embryogenesis, RIP1−/− FADD−/− double knockout (DKO) mice display perinatal lethality,15 similar to the phenotype of RIP1−/− single knockout mice.10 In contrast, deletion of a RIP1-related protein kinase, RIP3, fully restores normal embryonic as well as postnatal development in FADD−/− mice.21 Recent studies demonstrated that RIP1−/− mice can only reach adulthood when both FADD and RIP3 are absent, indicating that RIP1 protects neonatal cells from FADD-mediated apoptosis and RIP3-dependent necroptosis.22, 23, 24, 25 Importantly, FADD−/− RIP3−/− DKO mice and RIP1−/− FADD−/− RIP3−/− triple knockout mice develop age-dependent lymphadenopathy and splenomegaly, reminiscent of the lymphoproliferative (lpr) disease displayed by Fas−/− mice. Therefore, both apoptosis and necroptosis are essential for homeostasis in the peripheral lymphoid organs.Previous studies have indicated that RIP1 is essential for T-cell development, because RIP1-deficient fetal liver cells fail to reconstitute the T-cell compartment in immunodeficient recipient mice.15, 26 A recent study showed that lack of RIP1 in hematopoietic stem cells and progenitors (HSCs/Ps) leads to a severe defect in hematopoiesis.27 However, the temporal requirement for RIP1, particularly at postlineage commitment stages, remains unclear. In the current study, T lineage-specific deletion of RIP1 revealed a novel stage-specific requirement for RIP1 to protect T cells from apoptosis as well as to allow normal proliferative responses.  相似文献   

17.
Neuronal active Caspase-6 (Casp6) is associated with Alzheimer disease (AD), cognitive impairment, and axonal degeneration. Caspase-1 (Casp1) can activate Casp6 but the expression and functionality of Casp1-activating inflammasomes has not been well-defined in human neurons. Here, we show that primary cultures of human CNS neurons expressed functional Nod-like receptor protein 1 (NLRP1), absent in melanoma 2, and ICE protease activating factor, but not the NLRP3, inflammasome receptor components. NLRP1 neutralizing antibodies in a cell-free system, and NLRP1 siRNAs in neurons hampered stress-induced Casp1 activation. NLRP1 and Casp1 siRNAs also abolished stress-induced Casp6 activation in neurons. The functionality of the NLRP1 inflammasome in serum-deprived neurons was also demonstrated by NLRP1 siRNA-mediated inhibition of speck formation of the apoptosis-associated speck-like protein containing a caspase recruitment domain conjugated to green fluorescent protein. These results indicated a novel stress-induced intraneuronal NLRP1/Casp1/Casp6 pathway. Lipopolysaccharide induced Casp1 and Casp6 activation in wild-type mice brain cortex, but not in that of Nlrp1−/− and Casp1−/− mice. NLRP1 immunopositive neurons were increased 25- to 30-fold in AD brains compared with non-AD brains. NLRP1 immunoreactivity in these neurons co-localized with Casp6 activity. Furthermore, the NLRP1/Casp1/Casp6 pathway increased amyloid beta peptide 42 ratio in serum-deprived neurons. Therefore, CNS human neurons express functional NLRP1 inflammasomes, which activate Casp1 and subsequently Casp6, thus revealing a fundamental mechanism linking intraneuronal inflammasome activation to Casp1-generated interleukin-1-β-mediated neuroinflammation and Casp6-mediated axonal degeneration.The lack of efficient treatment for Alzheimer disease (AD) is of high social and economical cost and a growing concern with the aging of the world''s population.1 Therapies eliminating amyloid beta peptide (Aβ) from AD brains have unfortunately failed to stem progressive cognitive decline. These disappointing results have forced scientists to reconsider treatments against AD; some focusing on targeting Aβ earlier in disease, while others attempting to disaggregate the Tau protein in neurofibrillary tangles (NFT). Recently, the association of several immune responsive genes with increased AD risks2, 3, 4 have additionally revived interest in a possible etiological role for inflammation in AD.AD brain inflammation is attributed to activated microglia, which remove Aβ, and secrete neurotoxic molecules that induce neurodegeneration. Interleukin-1-beta (IL-1β), a critical component of brain neuroinflammation, is increased in AD brains5 and may contribute to AD pathology by increasing amyloid precursor protein (APP) gene expression, Tau hyperphosphorylation and memory impairment.6 However, anti-inflammatory therapies have not provided the expected beneficial effect in AD patients,7 suggesting that microglial inflammation may be a consequence of AD. Degenerating neurons are renowned initiators of brain inflammatory responses and the loss of synapses remains the best correlative marker of dementia in AD.8 This has incited us to study the response of human neurons to stress and to determine whether specific neuronal molecular events were initiated that link axonal degeneration to an inflammatory response.The active cysteinal Caspase-6 protease (Casp6), associated with axonal degeneration,9, 10, 11, 12, 13 is highly abundant in NFT, neuropil threads, and neuritic plaques of AD brains.14 In some aged non-cognitively impaired individuals, Casp6 activity in the entorhinal cortex and CA1 regions of the hippocampus,15 two areas initially affected by NFT pathology in AD,16 correlates significantly with lower cognitive performance.17 The expression of active Casp6 in CA1 pyramidal neurons of mouse brains is sufficient to induce age-dependent cognitive impairment, in the absence of plaques and tangles, which suggests that active Casp6 in AD brains could be a major contributor to axonal degeneration and cognitive decline.18Despite substantial evidence implicating Casp6 in AD, the pathways leading to Casp6 activation in neurons are unclear. Caspase-1 (Casp1) activates Casp6 in primary cultures of human CNS neurons.19 Inflammasome multiprotein complexes, constituted of danger sensing nucleotide-binding oligomerization domain-like receptors or the DNA sensing absent in melanoma 2 (AIM2) component, and the apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), recruit and induce Casp1 self-activation.20, 21 Functional Nod-like receptor protein 1 (NLRP1), Nod-like receptor protein 3 (NLRP3), AIM2, and ICE protease activating factor (IPAF-1) inflammasomes have been characterized primarily in peripheral macrophages22 and CNS microglia.23, 24 Recently, reports have indicated inflammasome receptor expression and activation in rodent neurons. Rat cerebellar granule neurons submitted to oxygen and glucose deprivation or reduced potassium levels increased Nlrp1 mRNA levels.25, 26 Nuclear Nlrp1 or functional Nlrp1 inflammasome complexes increased in rat cortical neurons after traumatic brain injury, stroke, and glucose-oxygen deprivation insults.27, 28, 29, 30, 31 Neuronal Nlrp1 increased in rats submitted to spinal cord or sciatic nerve injury,29, 32 and in aging rat hippocampus or ethanol treated hippocampal slice cultures.33, 34 Aim2 induced pyroptosis in rat cortical neuron cultures and traumatic brain injury.35 Nlrp1 has been reported in human brain pyramidal neurons36 and inflammasome receptor mRNAs were observed in human neuron cultures and human Rasmussen''s encephalitis.37Here, we assessed which inflammasome could activate Casp1 and subsequently Casp6 in human primary CNS cultures. We determined which inflammasomes were expressed in naive and stressed neurons and used siRNAs and S-100 cell-free extracts treated with specific inflammasome activators, or antibody blockers, to identify the functional inflammasome. We uncovered that the NLRP1, AIM2, and IPAF-1, but not the NLRP3, inflammasomes were expressed and functional in neurons and that the NLRP1 inflammasome was responsible for Casp1 and subsequently Casp6 activation in serum-deprived and benzylated ATP (BzATP)-stressed neurons. NLRP1 was co-localized with Casp6 activity, immunostained 25- to 30-fold more neurons in AD, and increased Aβ42 in serum-deprived neurons. The NLRP1–Casp1–Casp6 pathway was blocked in lipopolysaccharide (LPS)-treated Nlrp1/ and Casp1/ mice brains. These results reveal a molecular cascade linking neuronal inflammasome-mediated Casp1 activation to Casp6 activation and provide unexpected novel common neuronal therapeutic targets against neuroinflammation, axonal degeneration, and cognitive impairment in AD.  相似文献   

18.
The AMP-activated protein kinase, a key regulator of energy homeostasis, has a critical role in metabolic disorders and cancers. AMPK is mainly regulated by cellular AMP and phosphorylation by upstream kinases. Here, we show that PIKE-A binds to AMPK and blocks its tumor suppressive actions, which are mediated by tyrosine kinase Fyn. PIKE-A directly interacts with AMPK catalytic alpha subunit and impairs T172 phosphorylation, leading to repression of its kinase activity on the downstream targets. Mutation of Fyn phosphorylation sites on PIKE-A, depletion of Fyn, or pharmacological inhibition of Fyn blunts the association between PIKE-A and AMPK, resulting in loss of its inhibitory effect on AMPK. Cell proliferation and oncogenic assays demonstrate that PIKE-A antagonizes tumor suppressive actions of AMPK. In human glioblastoma samples, PIKE-A expression inversely correlates with the p-AMPK levels, supporting that PIKE-A negatively regulates AMPK activity in cancers. Thus, our findings provide additional layer of molecular regulation of the AMPK signaling pathway in cancer progression.AMP-activated protein kinase is activated under a variety of physiological and pathological stresses that increase the intracellular AMP/ATP ratio, either by increasing ATP consumption (exercise/muscle contraction) or by decreasing ATP production (e.g., glucose deprivation, hypoxia or ischemia). It is a heterotrimeric complex consisting of a catalytic α subunit and two regulatory (β and γ) subunits. An increase in intracellular AMP/ATP ratio results in allosteric activation of the kinase by protecting T172 from dephosphorylation.1 T172 phosphorylation in the activation loop of the α subunit is an absolute requirement for full activation of AMPK activity,2, 3 and is mediated by at least two distinct upstream kinases, liver kinase B1 (LKB1)4, 5, 6 and Ca2+/calmodulin-dependent kinase kinase β (CaMKKβ).7, 8, 9AMPK is an evolutionarily conserved metabolic sensor that has a pivotal role in maintaining energy homeostasis by coordinating metabolic pathways to balance nutrient supply and demand.10 Regulation of AMPK in multiple tissues is controlled by a growing number of hormones and cytokines, including leptin, adiponectin, IL-6, CNTF, TNF-α, and ghrelin. Moreover, AMPK can be activated by numerous small molecules such as metformin, aminoimidazole-4-carboxymide-1-β-D-ribofuranoside (AICAR), resveratrol, thiozolidinedione (TZD), and A-769662. Activated AMPK regulates glucose uptake and fatty acid oxidation in muscle and blocks gluconeogenesis in liver, enhancing insulin sensitivity. It also regulate appetite (for review, see Dzamko and Steinberg).11 In addition to these well-characterized functions in metabolic syndromes, AMPK serves as a metabolic tumor suppressor that reprograms the cellular metabolism and elicits a metabolic checkpoint on the cell cycle through its actions on mTORC1, p53, and other modulators for cell proliferation, cell growth, cell survival, and autophagy.12 Further, LKB1 activates AMPK and represses RNA synthesis.13 In LKB1-deficient lung cancer cells, AMPK activity is suppressed, leading to increased cell growth, whereas the ability of AMPK to inhibit cell growth is restored when wild-type LKB1 is expressed.14, 15 Additionally, the express levels of AMPK inversely correlate with clinical prognosis in gastric,16 breast, and ovarian tumors, and are diminished in cancer cells by activated PI3K pathways.17 Accumulating evidence supports that the susceptibility of cancer might be attributable to the dysregulated AMPK.18, 19 Hence, activation of AMPK may represent a novel target for cancer treatment.PIKE-A is a GTPase that directly interacts with PI 3 kinase or Akt and enhances their kinase activities.20, 21, 22, 23 It is a proto-oncogene that frequently amplified in numerous human cancers.24, 25 It binds Akt and escalates its kinase activity and promotes cancer cell survival, invasion, and migration.26, 27 Interestingly, PIKE knockout (PIKE−/−) mice are resistant to diet-induced obesity and diabetes,28 strongly implicating PIKE in obesity control. Accordingly, we observed higher AMPK phosphorylation and lipid oxidation in PIKE−/− muscle and fat tissues, which provide a mechanistic explanation to the slim phenotype of the knockout mice.28 Further, PIKE-A interacts with insulin receptor and mediates its suppressive effect on AMPK activation.29 Previously, we have reported that Fyn phosphorylates PIKE-A on both Y682 and Y77430 and regulates its interaction with different partners, promoting neuronal survival31 and adiposeness.32 In this report, we provide new evidence supporting that Fyn phosphorylation of PIKE-A is critical for its association with AMPK and inhibition of its kinase activity, leading to the blockade of cell proliferation. Hence, PIKE-A promotes tumorigenesis, at least, partially through blocking the tumor suppressive activity of AMPK. This discovery highlights a previously unappreciated relationship between cell metabolism and cell proliferation mediated by PIKE-A/AMPK complex.  相似文献   

19.
20.
Enhancing nitrogen use efficiency (NUE) in crop plants is an important breeding target to reduce excessive use of chemical fertilizers, with substantial benefits to farmers and the environment. In Arabidopsis (Arabidopsis thaliana), allocation of more NO3 to shoots was associated with higher NUE; however, the commonality of this process across plant species have not been sufficiently studied. Two Brassica napus genotypes were identified with high and low NUE. We found that activities of V-ATPase and V-PPase, the two tonoplast proton-pumps, were significantly lower in roots of the high-NUE genotype (Xiangyou15) than in the low-NUE genotype (814); and consequently, less vacuolar NO3 was retained in roots of Xiangyou15. Moreover, NO3 concentration in xylem sap, [15N] shoot:root (S:R) and [NO3] S:R ratios were significantly higher in Xiangyou15. BnNRT1.5 expression was higher in roots of Xiangyou15 compared with 814, while BnNRT1.8 expression was lower. In both B. napus treated with proton pump inhibitors or Arabidopsis mutants impaired in proton pump activity, vacuolar sequestration capacity (VSC) of NO3 in roots substantially decreased. Expression of NRT1.5 was up-regulated, but NRT1.8 was down-regulated, driving greater NO3 long-distance transport from roots to shoots. NUE in Arabidopsis mutants impaired in proton pumps was also significantly higher than in the wild type col-0. Taken together, these data suggest that decrease in VSC of NO3 in roots will enhance transport to shoot and essentially contribute to higher NUE by promoting NO3 allocation to aerial parts, likely through coordinated regulation of NRT1.5 and NRT1.8.China is the largest consumer of nitrogen (N) fertilizer in the world; however, the average N use efficiency (NUE) in fertilizer is only around 35%, suggesting considerable potential for improvements (Shen et al., 2003; Wang et al., 2014). With the high amounts of N-fertilizer being used, crop yields are declining in some areas, where application is exceeding the optimum required for local field crops (Shen et al., 2003; Miller and Smith, 2008; Xu et al., 2012). The extremely low NUE results in waste of resources and environmental contamination, and also presents serious hazards for human health (Xu et al., 2012; Chen et al., 2014). Consequently, exploiting the maximum potential for improving NUE in crop plants will have practical significance for agriculture production and the environment (Zhang et al., 2010; Schroeder et al., 2013; Wang et al., 2014). Elucidating the genetic and physiological regulatory mechanisms governing NUE in plants will allow breeding crops and varieties with higher NUE.Ammonium (NH4+) and nitrate (NO3) are the main N species absorbed and utilized by crops, and NO3 accumulation and utilization are of major emphasis for N nutrient studies in dry land crops, such as Brassica napus. Several studies revealed the close relationship between NO3 content and NUE in plant tissues (Shen et al., 2003; Zhang et al., 2012; Tang et al., 2013; Han et al., 2015a). When plants are sufficiently illuminated, NO3 assimilation efficiency significantly increase in shoots compared with roots (Smirnoff and Stewart, 1985; Tang et al., 2013). Consequently, under daytime with optimal illumination, higher proportion of NO3 in plant tissue is transported from root to shoot, as an advantageous physiological adaptation that reduces the cost of energy for metabolism (Tang et al., 2013). NO3 assimilation in plant shoots can therefore take advantage of solar energy while improving NUE (Smirnoff and Stewart, 1985; Andrews, 1986; Tang et al., 2012, 2013).The NO3 long-distance transport and distribution between root and shoot is regulated by two genes encoding long transport mechanisms. NRT1.5 is responsible for xylem NO3 loading, while NRT1.8 is responsible for xylem NO3 unloading (Lin et al., 2008; Li et al., 2010). Expression of the two genes is influenced by NO3 concentration. NRT1.5 is strongly induced by NO3 (Lin et al., 2008), while NRT1.8 expression is extremely up-regulated in nrt1.5 mutants (Chen et al., 2012). A negative correlation between the extents of expression of the two genes was observed when plants are subjected to abiotic stresses (Chen et al., 2012). Moreover, expression of NRT1.5 is strongly inhibited by 1-aminocyclopropane-1-carboxylic acid (ACC) and methyl jasmonate (MeJA), whereas the expression of NRT1.8 is significantly up-regulated (Zhang et al., 2014). Based on these studies, we argue that the expression and functioning of NO3 long-distance transport genes NRT1.5 and NRT1.8 are regulated by cytosolic NO3 concentration. In addition, the vacuolar and cytosolic NO3 distribution is likely regulated by proton pumps located within the tonoplast (V-ATPase and V-PPase; Granstedt and Huffaker, 1982; Glass et al., 2002; Krebs et al., 2010). Therefore, NO3 use efficiency must be affected by NO3 long-distant transport (between shoot and root) and short-distant transport (between vacuole and cytosol). However, the physiological mechanisms controlling this regulation are still obscure.Previous studies showed that the chloride channel protein (CLCa) is mainly responsible for vacuole NO3 short-distance transport, as it is the main channel for NO3 movement between the vacuoles and cytosol (De Angeli et al., 2006; Wege et al., 2014). The vacuole proton-pumps (V-ATPase and V-PPase) located in the tonoplast supply energy for active transport of NO3 and accumulation within the vacuole (Gaxiola et al., 2001; Brüx et al., 2008; Krebs et al., 2010). Despite the fact about 90% of the volume of mature plant cells is occupied by vacuoles, vacuolar NO3 cannot be efficiently assimilated because the enzyme nitrate reductase (NR) is cytosolic (Shen et al., 2003; Han et al., 2015a). However, retranslocation of NO3 from the vacuole to the cytosol will permit its immediate assimilation and utilization.Generally, NO3 concentrations in plant cell vacuoles and the cytoplasm are in the range of 30–50 mol m−3 and 3–5 mol m−3, respectively (Martinoia et al., 1981, 2000). Because vacuoles are obviously the organelle for high NO3 accumulation and storage in plant tissues, their function in NO3 use efficiency cannot be ignored (Martinoia et al., 1981; Zhang et al., 2012; Han et al., 2015b). NO3 assimilatory system in the cytoplasm is sufficient for its assimilation when it is transported out of the vacuoles. Therefore, NO3 use efficiency could in part be dependent on vacuolar-cytosolic NO3 short-distance transport in plant tissues (Martinoia et al., 1981; Shen et al., 2003; Zhang et al., 2012; Han et al., 2015a).Evidently, NO3 use efficiency is regulated by both NO3 long-distance transport from root to shoot and short-distance transport and distribution between vacuoles and cytoplasm within cells (Glass et al., 2002; Dechorgnat et al., 2011; Han et al., 2015a). Although vacuoles compartment excess NO3 that accumulates in plant cells (Granstedt and Huffaker, 1982; Krebs et al., 2010), neither NO3 inducible NR genes (NIA1 and NIA2; Fan et al., 2007; Han et al., 2015a) nor the NO3 long-distance transport gene NRT1.5 (Lin et al., 2008) are regulated by vacuolar NO3, even though they are essential for NO3 assimilation. Only NO3 transported from the vacuole to the cytosol can play a role in regulating NO3 inducible genes. Consequently, we argue that both NO3 assimilation in cells and its long-distance transport from root to shoot are regulated by cytosolic NO3 concentration. However, this hypothesis needs to be substantiated. The mechanisms underlying both NO3 short-distance (Gaxiola et al., 2001; De Angeli et al., 2006; Brüx et al., 2008; Krebs et al., 2010) and long-distance transport (Lin et al., 2008; Li et al., 2010) have been previously investigated, yet the underlying mechanisms regulating the flux of NO3 and the obvious relationship between the two transport pathways, as well as their relation to NUE, are not well understood.The NRT family of genes play a partial role in vacuolar NO3 accumulation in petioles (Chiu et al., 2004) and seed tissues (Chopin et al., 2007), whereas the proton pumps and CLCa system in the tonoplast play a major role in accumulating NO3 in vacuoles (Gaxiola et al., 2001; De Angeli et al., 2006; Brüx et al., 2008; Krebs et al., 2010). The vacuolar NO3 short-distance transport system is spread throughout the plant tissues and is the principal means by which vacuolar NO3 short-distance transport and distribution is controlled (De Angeli et al., 2006; Krebs et al., 2010).The NRT genes seem to work synergistically to control NO3 long-distance transport between roots and shoots. NRT1.9 is responsible for NO3 loading into the phloem (Wang and Tsay, 2011), whereas NO3 loading and unloading into xylem are regulated by NRT1.5 and NRT1.8, respectively (Lin et al., 2008; Li et al.; 2010). Phloem transport mainly involves organic N; the inorganic-N (NO3) concentrations in the phloem sap are typically very low, ranging from one-tenth to one-hundredth of that of the inorganic-N in xylem sap (Lin et al., 2008; Fan et al., 2009). Therefore, this study focused on NO3 short-distance transport mediated through the tonoplast proton pumps and the CLCa system and the long-distant transport mechanisms responsible for xylem NO3 loading and unloading via NRT1.5 and NRT1.8, respectively.Questions related to how long- and short-distance transport of NO3 are coupled in plant tissues and their role in determining NUE were addressed using a pair of high- and low-NUE B. napus genotypes and Arabidopsis (Arabidopsis thaliana). Application of proton pump inhibitors and ACC in the former, and use of mutants with defective proton pumps in the latter, allowed experimental distinction of the physiological mechanisms regulating these processes. Data presented here provide strong evidence from both model plants supporting this linkage and strongly suggest that cytosolic NO3 concentration in roots regulates NO3 long-distance transport from roots to shoots. We also investigated how NO3 concentration in plant tissues would be affected by NO3 long-distance transport, vacuolar NO3 sequestration, and the ensuing relationship with NO3 use efficiency. We also proposed the physiological mechanisms likely to be important for enhancing NO3 use efficiency in plants. These findings will provide scientific rationales for improving NUE in important industrial and food crops.  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