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 31 毫秒
1.
Infection with Helicobacter species is endemic in many animal facilities and may alter the penetrance of inflammatory bowel disease (IBD) phenotypes. However, little is known about the relative pathogenicity of H. typhlonius, H. rodentium, and combined infection in IBD models. We infected adult and neonatal IL10−/− mice with H. typhlonius, H. rodentium, or both bacteria. The severity of IBD and incidence of inflammation-associated colonic neoplasia were assessed in the presence and absence of antiHelicobacter therapy. Infected IL10−/− mice developed IBD with severity of noninfected (minimal to no inflammation) < H. rodentium < H. typhlonius < mixed H. rodentium + H. typhlonius (severe inflammation). Inflammation-associated colonic neoplasia was common in infected mice and its incidence correlated with IBD severity. Combined treatment with amoxicillin, clarithromycin, metronidazole, and omeprazole eradicated Helicobacter in infected mice and ameliorated established IBD in both infected and noninfected mice. Infection of IL10−/− mice with H. rodentium, H. typhlonius, or both organisms can trigger development of severe IBD that eventually leads to colonic neoplasia. The high incidence and multiplicity of neoplastic lesions in infected mice make this model well-suited for future research related to the development and chemoprevention of inflammation-associated colon cancer. The similar antiinflammatory effect of antibiotic therapy in Helicobacter-infected and -noninfected IL10−/− mice with colitis indicates that unidentified microbiota in addition to Helicobacter drive the inflammatory process in this model. This finding suggests a complex role for both Helicobacter and other intestinal microbiota in the onset and perpetuation of IBD in these susceptible hosts.Abbreviations: IBD, Inflammatory bowel diseaseInflammatory bowel disease (IBD) is hypothesized to develop due to aberrant immune responses induced by gut microbes.5 IBD does not occur in germ-free IL10−/− mice,2,15 indicating the importance of microorganisms as environmental triggers of intestinal inflammation. However, conventionally colonized or specific pathogen-free IL10−/− mice may develop colitis spontaneously2,32 or in response to specific triggers such as nonsteroidal antiinflammatory drugs3,14 or infections with certain bacteria.6,16,18 The normal lack of ongoing immune responses against bacteria in subjects without IBD has been attributed to the immunologic tolerance that specifically downregulates immune responses against antigens derived from these bacteria. Nevertheless, despite a large number of studies, no single bacterial type has fulfilled Koch postulates and been confirmed as a cause of IBD in animals or humans.Previous studies used fluorescence in situ hybridization with probes specific for bacterial 16S rRNA combined with conventional histologic techniques to study the relationships between various species of intestinal bacteria and the mucosa in mice and humans with IBD.33,34 Those studies showed that in normal mice, most bacterial groups are separated from the mucosal surface by either a mucus layer that excludes bacteria or, in the cecum and proximal colon, by an ‘interlaced’ layer that is composed of tightly packed bacteria. The interlaced or mucus layer thus limits the contact of the bulk of the enteric bacteria with the mucosal epithelium. In contrast, complex biofilms composed of multiple species of bacteria that were firmly adherent to the mucosal surface were identified in the majority of colon tissue samples collected from humans and mice with IBD.33,34 The presence of a biofilm abrogates the protective effects of the normal layer of mucus and can allow luminal bacterial antigens and toxins to reach the unshielded epithelial surface, where they can trigger cascades of host inflammatory responses. Situations that cause defects in the epithelial surface or degrade the protective qualities of mucus or the interlaced layer (or both) may allow contact of bacterial antigens and adjuvants with immune cells located in the lamina propria and lead to the generation of immune responses that result in IBD.34Helicobacter species are used frequently to model microbial triggers of colon inflammation, because they have previously been linked to the development of both IBD- and inflammation-associated neoplasia.11,21,29 Most studies have been performed by using Helicobacter hepaticus or H. bilis.20 However, H. typhlonius, H. rodentium, H. muridarum, H. ganmani, H. trogontum and other species8,12,17,29,35 can also be endemic in research animal facilities. The pathophysiologic effects of these less-common Helicobacter species are, for the most part, poorly investigated.Most rodent Helicobacter species are urease-negative and therefore preferentially colonize the intestine, but some species produce urease enzyme and can translocate to the liver or colonize the biliary system.13 H. typhlonius was shown to cause an enteric disease characterized by mucosal hyperplasia and associated inflammation in the cecum and colon in immunodeficient mice11,23 and IL10−/− mice.18 H. typhlonius is genetically related most closely to H. hepaticus, having only 2.36% difference in the 16S rRNA gene sequence, but H. typhlonius has a unique intervening sequence in this gene that makes it easily recognizable by PCR.9,12 Molecular detection of this pathogen with PCR is rapid, sensitive and allows the detection of the early phases of infection; further enhanced sensitivity is achieved with nested primers.22 One of the most important features of PCR is that it can be performed noninvasively on fecal pellets. Data regarding the pathogenetic mechanisms of H. rodentium are scarce.35,36 H. rodentium alone apparently does not cause hepatitis or enteritis in A/JCr or C.B-17/IcrCrl-scidBr mice; however, coinfection with H. hepaticus and H. rodentium was associated with augmented cecal gene expression and clinical diarrheal disease in immunodeficient mice compared with mice infected with H. hepaticus alone.23Previous reports demonstrated that H. typhlonius was capable of initiating colitis in adult IL10−/− mice.10,11 In those studies, colitis was relatively mild, with no development of inflammation-associated neoplasia. H. rodentium has been described to be nonpathogenic in adult wild-type mice but did enhance cytokine production in the cecum of mice also infected with H. hepaticus.23 We recently observed rapid onset of severe IBD and a high incidence of inflammation-associated neoplasia in IL10−/− mice that were coinfected with both H. typhlonius and H. rodentium as pups.16 The current study was undertaken to determine the relative roles of H. rodentium and H. typhlonius, individually and in combination, and age at infection in the development of colon inflammation and inflammation-associated neoplasia in IL10−/− mice. Novel features of our model include controlled infection of the combination of H. typhlonius and H. rodentium9 and infection of IL10−/− mice during the neonatal period.  相似文献   

2.
Infection of mouse colonies with Helicobacter spp. has become an increasing concern for the research community. Although Helicobacter infection may cause clinical disease, investigators may be unaware that their laboratory mice are infected because the pathology of Helicobacter species is host-dependent and may not be recognized clinically. The effects of Helicobacter infections are not limited to the gastrointestinal system and can affect reproduction, the development of cancers in gastrointestinal organs and remote organs such as the breast, responses to vaccines, and other areas of research. The data we present in this review show clearly that unintentional Helicobacter infection has the potential to significantly interfere with the reliability of research studies based on murine models. Therefore, frequent screening of rodent research colonies for Helicobacter spp. and the eradication of these pathogens should be key goals of the research community.The reliability of an experiment that uses an in vivo model system depends on understanding and controlling all variables that can influence the experimental outcome. Infections of mouse colonies are important to the scientific community because they can introduce such harmful variables. Therefore, the ultimate goal of laboratory animal facilities is to maintain disease-free animals, to eliminate those unwanted variables.Numerous pathogenic microbes can interfere with animal research (reviewed in reference 57), and colonization of mouse colonies with members of the family Helicobacteriaceae is an increasing concern for the research community. Naturally acquired Helicobacter infections have been reported in all commonly used laboratory rodent species.3,10,36,44,45,49,82,124 A study of mice derived from 34 commercial and academic institutions in Canada, Europe, Asia, Australia, and the United States showed that 88% of these institutions had mouse colonies infected with 1 or more Helicobacter spp.109 Approximately 59% of these mice were infected with Helicobacter hepaticus ; however monoinfections with other species also were encountered. In another study, at least 1 of 5 Helicobacter spp. was detected in 88% of the 40 mouse strains tested.4Surveys such as these have established that a broad range of Helicobacter spp. may be present in mouse research colonies. Several of those Helicobacter species cause disease in laboratory mice. H. hepaticus first was identified as a pathogen when it was discovered to be the cause of chronic hepatitis and hepatocellular carcinoma in mice,26,31,116 either alone or in combination with other Helicobacter spp.78 In addition, H. typhlonius causes intestinal inflammation in mice with immunodeficiency or defects in immune regulation;28,37 H. muridarum has been associated with gastritis,86 and H. bilis has been associated with hepatitis35,38 and colitis.60,61 Although, H. rodentium appears to be relatively nonpathogenic in wild-type and SCID mice,78 combined infection with H. rodentium and H. typhlonius results in a high incidence of inflammation-associated neoplasia in IL10−/− mice.9,46 Further, it is becoming increasingly clear that the effects of Helicobacter infections are not limited to the gastrointestinal system. Helicobacter infections have been documented to directly or indirectly affect responses as diverse as reproduction, development of breast cancer, and altered immune responses to vaccines.65,95,99 In addition to effects on rodents, Helicobacter spp. can infect other laboratory animals2,5,27,29,33,36,107 and can colonize different anatomic regions of the gastrointestinal system.35 This review focuses on the potential effect of these organisms on in vivo experiments and biomedical research. The results summarized here emphasize the importance of knowledge of colony infection status and prevention of unintentional infections to achieve the goal of providing a consistent and reliable environment for research studies.  相似文献   

3.
Enterohepatic Helicobacter species (EHS) often are associated with typhlocolitis and rectal prolapse in mice. We sought to describe rectal prolapses histologically, relate lesions to mouse genotype and EHS infection status, and characterize EHS pathogens on our campus. Our mouse population was housed among 6 facilities on our main campus and a seventh, nearby facility. We investigated cases of rectal prolapse over 1 y and included 76 mice, which were broadly categorized according to genotype. Microscopically, lesions ranged from mild to severe typhlocolitis, often with hyperplastic and dysplastic foci. Neoplastic foci tended to occur at the ileocecal–colic junction. Lesions were most severe in strains that had lower-bowel inflammatory disease, notably IL10, Rag1, and Rag2 knockout strains; prolapses occurred in these strains when housed both in areas with endemic EHS and in our Helicobacter-free barrier facility. Most mice with rectal prolapses were immunocompromised genetically modified mice; however, the most frequently sampled strain, the lamellipodin knockout, was noteworthy for its high incidence of rectal prolapse, localized distal colonic and rectal lesions, and lack of known immunodeficiency. This strain is being explored as a model of rectal carcinoma. Most of the colons examined tested PCR-positive for EHS, often with coinfections. Although H. bilis is prevalent on our campus, we did not find this organism in any mice exhibiting clinical signs of rectal prolapse. Identification of H. apodemus in 22% of cases has fueled increased surveillance on our campus to characterize this organism and differentiate it from the closely related H. rodentium.Abbreviations: EHS, enterohepatic Helicobacter species; IBD, inflammatory bowel disease; RFLP, restriction-fragment–length polymorphism; RP, rectal prolapseRectal prolapse (RP) occurs commonly in laboratory mice and is often associated with lower-bowel inflammation. Mice have a relatively short and poorly supported distal colon, which lacks a serosal covering.30 This anatomic weakness, coupled with a microbial insult, toxic injury, or space-occupying neoplastic masses within the gastrointestinal tract, are the predisposing factors for tenesmus and RP (Figure 1). In the context of microbial insults, the pathogenesis involves diffuse or multifocal inflammation in the more proximal segments of colon or distal colon, which can result in thickened edematous tissue and tenesmus, triggering a prolapse.6,30,40 Bacteria most often associated with this condition are the enterohepatic Helicobacter species (EHS) and Citrobacter rodentium; although in theory any pathogenic bacteria causing colitis may predispose mice to RP.1,11,13,38Open in a separate windowFigure 1.Mouse rectal prolapse. An example of the clinical presentation of rectal prolapse in laboratory mice. Note the attachment of bedding and nesting material in the film of mucous that frequently is seen covering the exposed rectal tissue. Generally the tissue becomes severely erythematous, as can be appreciated in this photograph.Although the clinical presentation of RP may occur in immunocompetent mice, it is most often associated with mice that have a spontaneous or transgenic mutation causing immunodeficiency.11,13,38 Indeed, these naturally occurring murine pathogens are used to model inflammatory bowel disease in strains that are highly susceptible to typhlocolitis with EHS infection; examples include Il10−/− and Rag-deficient mice.3,5,8,9,13,16,19,20,22,40 In addition, H. hepaticus and other EHS including H. typhlonius, H. rodentium, and H. bilis, which are known to persistently colonize the intestinal crypt of the lower bowel, have been shown to induce colitis-associated cancer in susceptible immunodeficient strains of mice.4,7,9,23,24,27,29,31In 1999, our institution introduced a rodent importation policy to reduce the introduction of murine pathogens. As part of this program, all approved commercial vendors were screened to ensure animals were SPF for EHS. Any random-source mice (typically imported from other academic institutions for collaborative projects) were required to be rederived by embryo transfer. In comparing PCR data between 1999 (prior to implementing the ET policy) and 2009, we found that after more than a decade of strict rederivation and husbandry practices that reduce fecal–oral transmission, EHS prevalence was markedly reduced.21 Despite this success, these practices did not completely eradicate rodent EHS. Of particular note, 2 facilities on campus house well-established long-term breeding colonies, many of which are unique transgenic lines with various immunodeficiencies, that are used primarily for immunology and cancer research. Rederivation of each of these strains was considered to be cost-prohibitive; thus EHS has remained endemic in these breeding colonies for more than a decade, as evident by our recent surveillance for EHS prevalence.21 The species known to be prevalent on our campus prior to the current study included H. hepaticus, H. rodentium, H. typhlonius, and H. bilis; in a few isolated areas, H. mastomyrinus was identified also.21Although EHS infections often are subclinical, we sought to correlate the presence of EHS-endemic areas with clinical lower-bowel inflammation (evident by rectal prolapse). In this survey of laboratory mice at our institution, we identified patterns in mouse strain susceptibility to RP, RP association with EHS, and histopathologic findings and correlated specific EHS species with clinical disease. Because we sought to study spontaneous infections, we excluded any mice on study with experimentally induced inflammatory bowel disease (IBD), including Helicobacter-induced IBD and chemically induced colitis models.From July 2011 to July 2012, a total of 63 mice with RP from these 6 facilities at our institution were necropsied as part of this investigation. In addition, 13 mice with RP were identified at a nearby research institute housing mice known to have endemic EHS.  相似文献   

4.
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   

5.
Murine norovirus (MNV) has recently been recognized as a widely prevalent viral pathogen in mouse colonies and causes disease and mortality in mice with impaired innate immunity. We tested the hypothesis that MNV infection would alter disease course and immune responses in mice with inflammatory bowel disease (IBD). FVB.129P2-Abcb1atm1Bor N7 (Mdr1a−/−) mice develop spontaneous IBD that is accelerated by infection with Helicobacter bilis. As compared with controls, Mdr1a−/− mice coinfected with MNV4 and H. bilis showed greater weight loss and IBD scores indicative of severe colitis, demonstrating that MNV4 can modulate the progression of IBD. Compared with controls, mice inoculated with MNV4 alone had altered levels of serum biomarkers, and flow cytometric analysis of immune cells from MNV4-infected mice showed changes in both dendritic cell (CD11c+) and other nonT cell (CD4 CD8) populations. Dendritic cells isolated from MNV4-infected mice induced higher IFNγ production by polyclonal T cells in vitro at 2 d after infection but not at later time points, indicating that MNV4 infection enhances antigen presentation by dendritic cells early after acute infection. These findings indicate that acute infection with MNV4 is immunomodulatory and alters disease progression in a mouse model of IBD.Abbreviations: DC, dendritic cell; IBD, inflammatory bowel disease; IP, IFNγ–inducible protein; MCP, macrophage chemotactic protein; MLN, mesenteric lymph node; MNV, murine norovirus; TNF, tumor necrosis factorThe genus Norovirus of the family Caliciviridae contains a large number of single-stranded, positive-sense RNA viruses that infect vertebrates, and strains have been identified in humans, cattle, swine, and (most recently) mice.19,29,34 Murine noroviruses (MNV) are recently recognized pathogens that can cause lethal infection in immunocompromised mice that lack innate immunity.19 However, MNV did not cause clinical disease in wild-type mice or many other strains of immunodeficient mice, including those lacking the recombination-activating gene (Rag−/−) and inducible nitric oxide synthase deficient mice.19,35,37 MNV was reported recently to be widespread in laboratory mice and may persist in immunocompetent animals, depending on the strain of MNV used.15,16,25 Studies in Rag−/− mice and B-cell–deficient strains showed that the acquired immune system plays an important role in the clearance of MNV.6,19,37 MNV has tropism for dendritic cells (DCs),36 which are important in the presentation of antigens to T cells in draining lymph nodes and in the pathogenesis of inflammatory bowel disease (IBD). Therefore, MNV is a potential confounder for in vivo immunology studies, including murine models of IBD.Idiopathic IBD, which encompasses both ulcerative colitis and Crohn disease, is a widely studied disorder that affects approximately 1.4 million people in the United States.20 Although the precise cause of human IBD has not been elucidated, studies with mouse models have demonstrated that abnormal host responses of the innate and adaptive immune systems to intestinal microbiota are important in the pathogenesis of IBD.28,38 DCs are the sentinels of the intestinal mucosal barrier and have a pivotal role in the initiation of IBD in response to microbial ligands.39 Alterations in DC responses could lead to persistence of bacterial infection, aberrant activation of the acquired immune system, and (ultimately) tissue damage.38Viral stimulation of DCs leads to activation of adaptive immune responses,17 including effector T cells, and as demonstrated with murine coronavirus (mouse hepatitis virus), intercurrent viral infections in mice can alter the phenotype of mouse models of human disease.10 Additional evidence suggests that intercurrent viral infection may enhance disease in human IBD patients.12,18 Whether infection with MNV alters DC function and, therefore, influences the progression of IBD in mouse models is unclear.Many mouse models of intestinal inflammation develop IBD that is driven by bacterial flora.9,28 Helicobacter spp. have been shown to drive this process in several mouse models including IL10-deficient, SMAD3-deficient, severe combined immunodeficiency and T-cell–deficient mice.4,5,13,23 FVB.129P2-Abcb1atm1Bor (Mdr1a−/−) mice develop spontaneous IBD that is accelerated by infection with Helicobacter bilis.21,22 In this report, we tested the hypothesis that infection with MNV can modulate IBD in this mouse model of bacterial-induced disease. We demonstrate that intercurrent MNV4 infection accelerates the progression of bacterial-induced IBD in the Mdr1a−/− mouse and alters the immune responses in this mouse model of IBD.  相似文献   

6.
Helicobacter spp. are some of the most prevalent bacterial contaminants of laboratory mice. Although abundant data regarding the diseases associated with H. hepaticus infection are available, little is known about the pathogenicity of H. ganmani, which was first isolated in 2001 from the intestines of laboratory mice. The objective of this study was to evaluate the host response to H. ganmani colonization in H. hepaticus disease-resistant C57BL/6 and disease-susceptible A/J and IL10-deficient mice. Mice were inoculated with H. ganmani, H. hepaticus, or Brucella broth. Cecal lesion scores, cecal gene expression, and Helicobacter load were measured at 4 and 90 d after inoculation. At both time points, mice inoculated with H. ganmani had similar or significantly more copies of cecum-associated Helicobacter DNA than did mice inoculated with H. hepaticus. When compared with those of sham-inoculated control mice, cecal lesion scores at 4 and 90 d after inoculation were not significantly greater in H. ganmani-inoculated A/J, C57BL/6, or IL10-deficient mice. Analysis of cecal gene expression demonstrated that H. ganmani infection failed to cause significant elevations of IFNγ in A/J, C57BL/6, or IL10-deficient mice. However, in IL10-deficient mice, H. ganmani infection was associated with a significant increase in the expression of the proinflammatory cytokine IL12/23p40. Although H. ganmani infection in this study failed to induce the typhlitis that is the hallmark of H. hepaticus infection, infection with H. ganmani was associated with alterations in inflammatory cytokines in IL10-deficient mice.Abbreviations: B6, C57BL/6NCr; HPRT, hypoxanthine guanine phosphoribosyl transferase; IL10 KO, B6129P2-IL10tm1Cgn/JSince the discovery of the link between Helicobacter pylori and chronic gastritis in 1982,17 Helicobacter spp. in humans and animals have become a field of extensive study. Due to improved detection methods, there has been a rapid expansion in our understanding and ability to detect native Helicobacter spp. in mouse models. Several reports investigating their prevalence in mice housed in research institutions have found Helicobacter spp. to be some of the most common bacterial contaminants of laboratory rodents.2,3,12,16,23 Helicobacter hepaticus is perhaps the most notorious of the murine helicobacters, by virtue of the early realization of its pathogenicity in adult mice.8,24 The hallmarks of infection by H. hepaticus are typhlitis, colitis, and hepatitis.10 In addition, H. hepaticus is commonly used as a microbial trigger in susceptible mouse strains used as models of inflammatory bowel disease.5,9,19,21,28 In 2001, less than 10 y after H. hepaticus was discovered, H. ganmani was isolated from the intestines of laboratory mice.26 During its initial characterization, 16S rDNA sequence analysis placed H. ganmani phylogenetically closest to H. rodentium, a urease-negative helicobacter that had been previously isolated from mouse intestines.26Despite the reported endemic presence of H. ganmani in many research colonies,2,3,12 only a few reports to date have attempted to address H. ganmani’s potential pathogenicity.22,30 One report describes an outbreak of inflammatory bowel-like disease associated with H. ganmani infection in an otherwise Helicobacter-free conventional colony of IL10-deficient mice.22 The findings from another report describe the effect of natural colonization of IL10-deficient mice with H. ganmani, H. hepaticus, or both.30 In that study, 8- to 20-wk-old mice monoinfected with H. ganmani had significantly lower lesion scores than did mice monoinfected with H. hepaticus, suggesting that infection with H. ganmani alone was not sufficient to cause severe typhlocolitis.30 However, by 34 wk of age, clinical typhlocolitis (diarrhea) and grossly enlarged ceca were observed at necropsy in 2 of the 6 mice monoinfected with H. ganmani.30Although these reports of naturally occurring infections have provided a glimpse into H. ganmani’s potential to produce intestinal disease in immunodeficient mice, a controlled study in immunocompetent and immunodeficient mice had not been conducted previously. The objectives of the current study were to evaluate the effect of H. ganmani infection on intestinal disease and to characterize alterations of inflammatory gene expression associated with infection. To this end, we selected A/J and IL10-deficient mice for this study because of their known susceptibility to H. hepaticus-induced typhlocolitis.9,13,14,19,21,28 In contrast, although C57BL/6 mice show an initial spike in inflammatory cytokines after H. hepaticus infection, they do not typically develop chronic disease.19 We did not expect C57BL/6 mice to develop H. hepaticus-induced disease, but we deemed it prudent to characterize the possible effects—through unknown mechanisms—of H. ganmani on this common strain.Previous studies characterizing cecal gene expression during H. hepaticus induced typhlocolitis demonstrated that IFNγ and IL12/23p40 (IL12/23) are key proinflammatory cytokines that drive typhlitis.19 Expression of these cytokines was increased in H. hepaticus-inoculated A/J mice but not in H. hepaticus-inoculated C57BL/6 mice.19 In addition, treatment with neutralizing monoclonal antibodies against these cytokines significantly decreased cecal lesion severity, implicating the roles of IFNγ and IL12/23 in modulating the pathogenesis of typhlitis.19 We hypothesized that characterizing the effect of H. ganmani infection on expression of IFNγ and IL12/23 would uncover aspects of the host response that are not readily apparent by histologic evaluation of cecal tissue alone.To date, our understanding of the potential for H. ganmani to cause intestinal disease has been limited to reports that focused on the evaluation of histologic disease in naturally infected IL10-deficient mice. Despite the reported endemic presence of H. ganmani in many research colonies,2,3,12 there are no published reports of disease associated with H. ganmani infection in immunocompetent mice. In addition, H. ganmani shares close sequence homology with H. rodentium, which has been found to be nonpathogenic in monoinfected immunodeficient and immunocompetent mice.20 Therefore, we hypothesized that experimental infection with H. ganmani would not produce disease in H. hepaticus-susceptible or -resistant mice.  相似文献   

7.
8.
9.
10.
11.
12.
13.
14.
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   

15.
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   

16.
17.
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   

18.
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   

19.
20.
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   

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