Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Methods Protoc ; 6(2)2023 Apr 07.
Article in English | MEDLINE | ID: mdl-37104024

ABSTRACT

Outer membrane vesicles (OMVs) are lipid structures containing various biomolecules in their native environment and are spontaneously shed by gram-negative bacteria. OMVs perform several biological functions critical to both bacterial physiology and pathogenicity. Scientific research on OMV function and biogenesis requires a standardized and robust method of isolating these vesicles from bacterial cultures that reliably provide high-purity OMVs. Herein, we describe an optimized protocol to isolate OMVs from overnight cultures of three different strains of nontypeable Haemophilus influenzae (NTHi) for use in different downstream applications. Involving mainly differential centrifugation of the culture supernatant, the procedure described is relatively simple, efficient, and generates high-quality OMV preparations from each strain tested with sufficient yields, while preserving the native outer membrane composition.

2.
Bone Marrow Transplant ; 55(6): 1059-1067, 2020 06.
Article in English | MEDLINE | ID: mdl-31898692

ABSTRACT

Due to the curative potential and improvement in progression-free survival (PFS), high-dose chemotherapy followed by autologous stem cell transplantation (ASCT) is considered the standard of care for several hematologic malignancies, such as multiple myeloma, and lymphomas. ASCT typically involves support with blood product transfusion. Thus, difficulties arise when Jehovah's Witness patients refuse blood transfusions. In order to demonstrate the safety of performing "bloodless" ASCT (BL-ASCT), we performed a retrospective analysis of 66 Jehovah's Witnesses patients who underwent BL-ASCT and 1114 non-Jehovah's Witness patients who underwent transfusion-supported ASCT (TF-ASCT) at Cedars-Sinai Medical Center between January 2000 and September 2018. Survival was compared between the two groups. Transplant-related complications, mortality, engraftment time, length of hospital stay, and number of ICU transfers were characterized for the BL-ASCT group. One year survival was found to be 87.9% for both groups (P = 0.92). In the BL-ASCT group, there was one death prior to the 30 days post transplant due to CNS hemorrhage, and one death prior to 100 days due to sepsis. Based on our data, BL-ASCT can be safely performed with appropriate supportive measures, and we encourage community oncologists to promptly refer JW patients for transplant evaluation when ASCT is indicated.


Subject(s)
Hematopoietic Stem Cell Transplantation , Jehovah's Witnesses , Blood Transfusion , Humans , Retrospective Studies , Transplantation, Autologous
4.
J Biol Chem ; 292(42): 17290-17301, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28842504

ABSTRACT

Clostridium difficile is a major nosocomial pathogen that produces two exotoxins, TcdA and TcdB, with TcdB thought to be the primary determinant in human disease. TcdA and TcdB are large, multidomain proteins, each harboring a cytotoxic glucosyltransferase domain that is delivered into the cytosol from endosomes via a translocation domain after receptor-mediated endocytosis of toxins from the cell surface. Although there are currently no known host cell receptors for TcdA, three cell-surface receptors for TcdB have been identified: CSPG4, NECTIN3, and FZD1/2/7. The sites on TcdB that mediate binding to each receptor are not defined. Furthermore, it is not known whether the combined repetitive oligopeptide (CROP) domain is involved in or required for receptor binding. Here, in a screen designed to identify sites in TcdB that are essential for target cell intoxication, we identified a region at the junction of the translocation and the CROP domains that is implicated in CSPG4 binding. Using a series of C-terminal truncations, we show that the CSPG4-binding site on TcdB extends into the CROP domain, requiring three short repeats for binding and for full toxicity on CSPG4-expressing cells. Consistent with the location of the CSPG4-binding site on TcdB, we show that the anti-TcdB antibody bezlotoxumab, which binds partially within the first three short repeats, prevents CSPG4 binding to TcdB. In addition to establishing the binding region for CSPG4, this work ascribes for the first time a role in TcdB CROPs in receptor binding and further clarifies the relative roles of host receptors in TcdB pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Clostridioides difficile/enzymology , Glucosyltransferases/metabolism , Membrane Proteins/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/genetics , Broadly Neutralizing Antibodies , CHO Cells , Caco-2 Cells , Chlorocebus aethiops , Chondroitin Sulfate Proteoglycans/genetics , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Cricetinae , Cricetulus , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Protein Binding , Protein Domains
5.
J Mol Biol ; 429(7): 1030-1044, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28232034

ABSTRACT

The exotoxins toxin A (TcdA) and toxin B (TcdB) are produced by the bacterial pathogen Clostridium difficile and are responsible for the pathology associated with C. difficile infection (CDI). The antitoxin antibodies actoxumab and bezlotoxumab bind to and neutralize TcdA and TcdB, respectively. Bezlotoxumab was recently approved by the FDA for reducing the recurrence of CDI. We have previously shown that a single molecule of bezlotoxumab binds to two distinct epitopes within the TcdB combined repetitive oligopeptide (CROP) domain, preventing toxin binding to host cells. In this study, we characterize the binding of actoxumab to TcdA and examine its mechanism of toxin neutralization. Using a combination of approaches including a number of biophysical techniques, we show that there are two distinct actoxumab binding sites within the CROP domain of TcdA centered on identical amino acid sequences at residues 2162-2189 and 2410-2437. Actoxumab binding caused the aggregation of TcdA especially at higher antibody:toxin concentration ratios. Actoxumab prevented the association of TcdA with target cells demonstrating that actoxumab neutralizes toxin activity by inhibiting the first step of the intoxication cascade. This mechanism of neutralization is similar to that observed with bezlotoxumab and TcdB. Comparisons of the putative TcdA epitope sequences across several C. difficile ribotypes and homologous repeat sequences within TcdA suggest a structural basis for observed differences in actoxumab binding and/or neutralization potency. These data provide a mechanistic basis for the protective effects of the antibody in vitro and in vivo, including in various preclinical models of CDI.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Bacterial Toxins/antagonists & inhibitors , Enterotoxins/antagonists & inhibitors , Epitopes/metabolism , Binding Sites , Broadly Neutralizing Antibodies , Protein Aggregates , Protein Binding
6.
Antimicrob Agents Chemother ; 60(11): 6471-6482, 2016 11.
Article in English | MEDLINE | ID: mdl-27527088

ABSTRACT

Clostridium difficile causes infections of the colon in susceptible patients. Specifically, gut dysbiosis induced by treatment with broad-spectrum antibiotics facilitates germination of ingested C. difficile spores, expansion of vegetative cells, and production of symptom-causing toxins TcdA and TcdB. The current standard of care for C. difficile infections (CDI) consists of administration of antibiotics such as vancomycin that target the bacterium but also perpetuate gut dysbiosis, often leading to disease recurrence. The monoclonal antitoxin antibodies actoxumab (anti-TcdA) and bezlotoxumab (anti-TcdB) are currently in development for the prevention of recurrent CDI. In this study, the effects of vancomycin or actoxumab/bezlotoxumab treatment on progression and resolution of CDI were assessed in mice and hamsters. Rodent models of CDI are characterized by an early severe phase of symptomatic disease, associated with high rates of morbidity and mortality; high intestinal C. difficile burden; and a disrupted intestinal microbiota. This is followed in surviving animals by gradual recovery of the gut microbiota, associated with clearance of C. difficile and resolution of disease symptoms over time. Treatment with vancomycin prevents disease initially by inhibiting outgrowth of C. difficile but also delays microbiota recovery, leading to disease relapse following discontinuation of therapy. In contrast, actoxumab/bezlotoxumab treatment does not impact the C. difficile burden but rather prevents the appearance of toxin-dependent symptoms during the early severe phase of disease, effectively preventing disease until the microbiota (the body's natural defense against C. difficile) has fully recovered. These data provide insight into the mechanism of recurrence following vancomycin administration and into the mechanism of recurrence prevention observed clinically with actoxumab/bezlotoxumab.


Subject(s)
Anti-Bacterial Agents/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antitoxins/pharmacology , Clostridium Infections/drug therapy , Vancomycin/adverse effects , Animals , Anti-Bacterial Agents/administration & dosage , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/biosynthesis , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/biosynthesis , Broadly Neutralizing Antibodies , Clostridioides difficile/drug effects , Clostridioides difficile/growth & development , Clostridioides difficile/pathogenicity , Clostridium Infections/immunology , Clostridium Infections/microbiology , Clostridium Infections/mortality , Convalescence , Cricetulus , Disease Models, Animal , Disease Progression , Enterotoxins/antagonists & inhibitors , Enterotoxins/biosynthesis , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Humans , Mice , Mice, Inbred C57BL , Survival Analysis , Vancomycin/administration & dosage
7.
Int J Endocrinol ; 2015: 401851, 2015.
Article in English | MEDLINE | ID: mdl-26448747

ABSTRACT

African American women suffer higher incidence and mortality of triple negative breast cancer (TNBC) than Caucasian women. TNBC is very aggressive, causing the worst clinical outcome. We previously demonstrated that tumors from these patients express high IGF-II and exhibit high activation of the IGF signaling pathways. IGF-II gene expression is imprinted (monoallelic), promotes tumor progression, and metastasis and regulates Survivin, a TNBC prognostic marker. Since BC mortality has increased among young Vietnamese women, we analyzed 48 (paired) TNBC samples from Vietnamese patients to assess IGF-II expression. We analyzed all samples by qrtPCR for identification of IGF-II heterozygosity and to determine allelic expression of the IGF-II gene. We also analyzed the tissues for proIGF-II and Survivin by RT-PCR and Western blotting. A total of 28 samples displayed IGF-II heterozygosity of which 78% were biallelic. Tumors with biallelic IGF-II gene expression exhibited the highest levels of proIGF-II and Survivin. Although 100% of these tissues corresponding normal samples were biallelic, they expressed significantly lower levels of or no proIGF-II and Survivin. Thus, IGF-II biallelic gene expression is differentially regulated in normal versus tumor tissues. We propose that intratumoral proIGF-II is dependent on the IGF-II gene imprinting status and it will promote a more aggressive TNBC.

8.
Antimicrob Agents Chemother ; 59(2): 1052-60, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25451052

ABSTRACT

Clostridium difficile infections (CDIs) are the leading cause of hospital-acquired infectious diarrhea and primarily involve two exotoxins, TcdA and TcdB. Actoxumab and bezlotoxumab are human monoclonal antibodies that neutralize the cytotoxic/cytopathic effects of TcdA and TcdB, respectively. In a phase II clinical study, the actoxumab-bezlotoxumab combination reduced the rate of CDI recurrence in patients who were also treated with standard-of-care antibiotics. However, it is not known whether the antibody combination will be effective against a broad range of C. difficile strains. As a first step toward addressing this, we tested the ability of actoxumab and bezlotoxumab to neutralize the activities of toxins from a number of clinically relevant and geographically diverse strains of C. difficile. Neutralization potencies, as measured in a cell growth/survival assay with purified toxins from various C. difficile strains, correlated well with antibody/toxin binding affinities. Actoxumab and bezlotoxumab neutralized toxins from culture supernatants of all clinical isolates tested, including multiple isolates of the BI/NAP1/027 and BK/NAP7/078 strains, at antibody concentrations well below plasma levels observed in humans. We compared the bezlotoxumab epitopes in the TcdB receptor binding domain across known TcdB sequences and found that key substitutions within the bezlotoxumab epitopes correlated with the relative differences in potencies of bezlotoxumab against TcdB of some strains, including ribotypes 027 and 078. Combined with in vitro neutralization data, epitope modeling will enhance our ability to predict the coverage of new and emerging strains by actoxumab-bezlotoxumab in the clinic.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Clostridioides difficile/drug effects , Bacterial Proteins/genetics , Cell Line , Clostridioides difficile/immunology , Clostridioides difficile/pathogenicity , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Epitopes/immunology , Female , Humans , Male
9.
Infect Immun ; 83(2): 822-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25486992

ABSTRACT

Clostridium difficile infection (CDI) represents the most prevalent cause of antibiotic-associated gastrointestinal infections in health care facilities in the developed world. Disease symptoms are caused by the two homologous exotoxins, TcdA and TcdB. Standard therapy for CDI involves administration of antibiotics that are associated with a high rate of disease recurrence, highlighting the need for novel treatment paradigms that target the toxins rather than the organism itself. A combination of human monoclonal antibodies, actoxumab and bezlotoxumab, directed against TcdA and TcdB, respectively, has been shown to decrease the rate of recurrence in patients treated with standard-of-care antibiotics. However, the exact mechanism of antibody-mediated protection is poorly understood. In this study, we show that the antitoxin antibodies are protective in multiple murine models of CDI, including systemic and local (gut) toxin challenge models, as well as primary and recurrent models of infection in mice. Systemically administered actoxumab-bezlotoxumab prevents both the damage to the gut wall and the inflammatory response, which are associated with C. difficile in these models, including in mice challenged with a strain of the hypervirulent ribotype 027. Furthermore, mutant antibodies (N297Q) that do not bind to Fcγ receptors provide a level of protection similar to that of wild-type antibodies, demonstrating that the mechanism of protection is through direct neutralization of the toxins and does not involve host effector functions. These data provide a mechanistic basis for the prevention of recurrent disease observed in CDI patients in clinical trials.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antitoxins/immunology , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/prevention & control , Enterotoxins/immunology , Animals , Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Antitoxins/therapeutic use , Chlorocebus aethiops , Disease Models, Animal , Enterocolitis, Pseudomembranous/immunology , Mice , Mice, Inbred C57BL , Mutation , Receptors, IgG/immunology , Recurrence , Vero Cells
10.
J Biol Chem ; 289(26): 18008-21, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24821719

ABSTRACT

The symptoms of Clostridium difficile infections are caused by two exotoxins, TcdA and TcdB, which target host colonocytes by binding to unknown cell surface receptors, at least in part via their combined repetitive oligopeptide (CROP) domains. A combination of the anti-TcdA antibody actoxumab and the anti-TcdB antibody bezlotoxumab is currently under development for the prevention of recurrent C. difficile infections. We demonstrate here through various biophysical approaches that bezlotoxumab binds to specific regions within the N-terminal half of the TcdB CROP domain. Based on this information, we solved the x-ray structure of the N-terminal half of the TcdB CROP domain bound to Fab fragments of bezlotoxumab. The structure reveals that the TcdB CROP domain adopts a ß-solenoid fold consisting of long and short repeats and that bezlotoxumab binds to two homologous sites within the CROP domain, partially occluding two of the four putative carbohydrate binding pockets located in TcdB. We also show that bezlotoxumab neutralizes TcdB by blocking binding of TcdB to mammalian cells. Overall, our data are consistent with a model wherein a single molecule of bezlotoxumab neutralizes TcdB by binding via its two Fab regions to two epitopes within the N-terminal half of the TcdB CROP domain, partially blocking the carbohydrate binding pockets of the toxin and preventing toxin binding to host cells.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Clostridioides difficile/immunology , Epitopes/immunology , Amino Acid Sequence , Antibodies, Bacterial/chemistry , Antibodies, Monoclonal , Antibodies, Neutralizing/chemistry , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Binding Sites , Broadly Neutralizing Antibodies , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , Crystallography, X-Ray , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary
11.
Prev Chronic Dis ; 9: E35, 2012.
Article in English | MEDLINE | ID: mdl-22239750

ABSTRACT

BACKGROUND: To address cardiovascular disease risk factors among Hispanics, a community model of prevention requires a comprehensive approach to community engagement. The objectives of our intervention were to reduce cardiovascular disease risk factors in Hispanics living in 2 low-income areas of El Paso, Texas, and to engage the community in a physical activity and nutrition intervention. METHODS: Drawing on lessons learned in phase 1 (years 2005-2008) of the HEART Project, we used an iterative, community-based process to develop an intervention based on an ecological framework. New community partners were introduced and community health workers delivered several elements of the intervention, including the curriculum entitled "Mi Corazón, Mi Comunidad" ("MiCMiC" [My Heart, My Community]). We received feedback from the project's Community Health Academy and Leadership Council throughout the development process and established a policy agenda that promotes integration of community health workers into the local and state workforce. OUTCOME: Collaboration with 2 new community partners, the YWCA and the Department of Parks and Recreation, were instrumental in the process of community-based participatory research. We enrolled 113 participants in the first cohort; 78% were female, and the mean age was 41 years. More than 50% reported having no health insurance coverage. Seventy-two (60%) participants attended 1 or more promotora-led Su Corazón, Su Vida sessions, and 74 (62%) participants attended 1 or more of the 15 exercise classes. INTERPRETATION: HEART phase 2 includes a multilevel ecological model to address cardiovascular disease risk among Hispanics. Future similarly targeted initiatives can benefit from an ecological approach that also embraces the promotora model.


Subject(s)
Cardiovascular Diseases/prevention & control , Community Health Services/methods , Community-Based Participatory Research/methods , Health Education/organization & administration , Health Promotion , Program Evaluation , Public Health , Adult , Female , Humans , Male , Mexico , Risk Factors , Texas
12.
Proc Natl Acad Sci U S A ; 104(18): 7612-6, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17456595

ABSTRACT

Emergence of bacterial resistance is a major issue for all classes of antibiotics; therefore, the identification of new classes is critically needed. Recently we reported the discovery of platensimycin by screening natural product extracts using a target-based whole-cell strategy with antisense silencing technology in concert with cell free biochemical validations. Continued screening efforts led to the discovery of platencin, a novel natural product that is chemically and biologically related but different from platensimycin. Platencin exhibits a broad-spectrum Gram-positive antibacterial activity through inhibition of fatty acid biosynthesis. It does not exhibit cross-resistance to key antibiotic resistant strains tested, including methicillin-resistant Staphylococcus aureus, vancomycin-intermediate S. aureus, and vancomycin-resistant Enterococci. Platencin shows potent in vivo efficacy without any observed toxicity. It targets two essential proteins, beta-ketoacyl-[acyl carrier protein (ACP)] synthase II (FabF) and III (FabH) with IC50 values of 1.95 and 3.91 microg/ml, respectively, whereas platensimycin targets only FabF (IC50 = 0.13 microg/ml) in S. aureus, emphasizing the fact that more antibiotics with novel structures and new modes of action can be discovered by using this antisense differential sensitivity whole-cell screening paradigm.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/antagonists & inhibitors , Aminophenols/pharmacology , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Polycyclic Compounds/pharmacology , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Aminophenols/chemistry , Anti-Bacterial Agents/chemistry , Enzyme Inhibitors/chemistry , Microbial Viability/drug effects , Molecular Structure , Polycyclic Compounds/chemistry
13.
Nature ; 441(7091): 358-61, 2006 May 18.
Article in English | MEDLINE | ID: mdl-16710421

ABSTRACT

Bacterial infection remains a serious threat to human lives because of emerging resistance to existing antibiotics. Although the scientific community has avidly pursued the discovery of new antibiotics that interact with new targets, these efforts have met with limited success since the early 1960s. Here we report the discovery of platensimycin, a previously unknown class of antibiotics produced by Streptomyces platensis. Platensimycin demonstrates strong, broad-spectrum Gram-positive antibacterial activity by selectively inhibiting cellular lipid biosynthesis. We show that this anti-bacterial effect is exerted through the selective targeting of beta-ketoacyl-(acyl-carrier-protein (ACP)) synthase I/II (FabF/B) in the synthetic pathway of fatty acids. Direct binding assays show that platensimycin interacts specifically with the acyl-enzyme intermediate of the target protein, and X-ray crystallographic studies reveal that a specific conformational change that occurs on acylation must take place before the inhibitor can bind. Treatment with platensimycin eradicates Staphylococcus aureus infection in mice. Because of its unique mode of action, platensimycin shows no cross-resistance to other key antibiotic-resistant strains tested, including methicillin-resistant S. aureus, vancomycin-intermediate S. aureus and vancomycin-resistant enterococci. Platensimycin is the most potent inhibitor reported for the FabF/B condensing enzymes, and is the only inhibitor of these targets that shows broad-spectrum activity, in vivo efficacy and no observed toxicity.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/antagonists & inhibitors , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Acetamides/pharmacology , Acetamides/toxicity , Adamantane , Aminobenzoates , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Aminoglycosides/toxicity , Anilides , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/toxicity , Apoproteins/chemistry , Apoproteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Linezolid , Lipids/biosynthesis , Mice , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Oxazolidinones/pharmacology , Oxazolidinones/toxicity , Streptomyces/metabolism , Substrate Specificity
14.
Science ; 304(5678): 1805-7, 2004 Jun 18.
Article in English | MEDLINE | ID: mdl-15205533

ABSTRACT

Salmonella enterica, the cause of food poisoning and typhoid fever, induces actin cytoskeleton rearrangements and membrane ruffling to gain access into nonphagocytic cells, where it can replicate and avoid innate immune defenses. Here, we found that SopB, a phosphoinositide phosphatase that is delivered into host cells by a type III secretion system, was essential for the establishment of Salmonella's intracellular replicative niche. SopB mediated the formation of spacious phagosomes following bacterial entry and was responsible for maintaining high levels of phosphatidylinositol-three-phosphate [PtdIns(3)P] in the membrane of the bacteria-containing vacuoles. Absence of SopB caused a significant defect in the maturation of the Salmonella-containing vacuole and impaired bacterial intracellular growth.


Subject(s)
Bacterial Proteins/metabolism , Cytoplasmic Vesicles/microbiology , Intestinal Mucosa/microbiology , Phagosomes/microbiology , Phosphatidylinositols/metabolism , Salmonella typhimurium/metabolism , Antigens, CD/metabolism , Bacterial Proteins/genetics , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Epithelial Cells/microbiology , Gene Deletion , Genomic Islands , Humans , Intestinal Mucosa/cytology , Lysosomal Membrane Proteins , Microscopy, Video , Mutation , Phagosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Recombinant Fusion Proteins/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development , Salmonella typhimurium/pathogenicity , Vacuoles/metabolism , Vacuoles/microbiology , Vacuoles/ultrastructure
15.
J Cell Biol ; 163(5): 1123-31, 2003 Dec 08.
Article in English | MEDLINE | ID: mdl-14662750

ABSTRACT

Salmonella enterica, the causative agent of food poisoning and typhoid fever, induces programmed cell death in macrophages, a process found to be dependent on a type III protein secretion system, and SipB, a protein with membrane fusion activity that is delivered into host cells by this system. When expressed in cultured cells, SipB caused the formation of and localized to unusual multimembrane structures. These structures resembled autophagosomes and contained both mitochondrial and endoplasmic reticulum markers. A mutant form of SipB devoid of membrane fusion activity localized to mitochondria, but did not induce the formation of membrane structures. Upon Salmonella infection of macrophages, SipB was found in mitochondria, which appeared swollen and devoid of christae. Salmonella-infected macrophages exhibited marked accumulation of autophagic vesicles. We propose that Salmonella, through the action of SipB, kills macrophages by disrupting mitochondria, thereby inducing autophagy and cell death.


Subject(s)
Autophagy/physiology , Bacterial Proteins/metabolism , Cell Death , Macrophages/physiology , Salmonella typhimurium/metabolism , Animals , Bacterial Proteins/genetics , COS Cells , Caspase 1/genetics , Caspase 1/metabolism , Endoplasmic Reticulum/metabolism , Macrophages/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Salmonella typhimurium/pathogenicity
17.
Cell ; 110(2): 191-202, 2002 Jul 26.
Article in English | MEDLINE | ID: mdl-12150927

ABSTRACT

Toll-like receptors (TLRs) detect microorganisms and protect multicellular organisms from infection. TLRs transduce their signals through MyD88 and the serine/threonine kinase IRAK. The IRAK family consists of two active kinases, IRAK and IRAK-4, and two inactive kinases, IRAK-2 and IRAK-M. IRAK-M expression is restricted to monocytes/macrophages, whereas other IRAKs are ubiquitous. We show here that IRAK-M is induced upon TLR stimulation and negatively regulates TLR signaling. IRAK-M prevented dissociation of IRAK and IRAK-4 from MyD88 and formation of IRAK-TRAF6 complexes. IRAK-M(-/-) cells exhibited increased cytokine production upon TLR/IL-1 stimulation and bacterial challenge, and IRAK-M(-/-) mice showed increased inflammatory responses to bacterial infection. Endotoxin tolerance, a protection mechanism against endotoxin shock, was significantly reduced in IRAK-M(-/-) cells. Thus, IRAK-M regulates TLR signaling and innate immune homeostasis.


Subject(s)
Drosophila Proteins , Membrane Glycoproteins/immunology , Protein Kinases/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing , Animals , Antigens, Differentiation/biosynthesis , Base Sequence , Cell Line, Transformed , Cells, Cultured , Cloning, Molecular , DNA, Complementary , Escherichia coli/immunology , Female , Humans , Interleukin-1/pharmacology , Interleukin-1 Receptor-Associated Kinases , Interleukin-12/biosynthesis , Interleukin-6/biosynthesis , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Listeria monocytogenes , Macrophages/cytology , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/immunology , Molecular Sequence Data , Myeloid Differentiation Factor 88 , NF-kappa B/immunology , Protein Biosynthesis , Protein Kinases/genetics , Receptors, Immunologic/biosynthesis , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella typhimurium/immunology , TNF Receptor-Associated Factor 6 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases
18.
Nature ; 416(6877): 194-9, 2002 Mar 14.
Article in English | MEDLINE | ID: mdl-11894098

ABSTRACT

The immune system consists of two evolutionarily different but closely related responses, innate immunity and adaptive immunity. Each of these responses has characteristic receptors-Toll-like receptors (TLRs) for innate immunity and antigen-specific receptors for adaptive immunity. Here we show that the caspase recruitment domain (CARD)-containing serine/threonine kinase Rip2 (also known as RICK, CARDIAK, CCK and Ripk2) transduces signals from receptors of both immune responses. Rip2 was recruited to TLR2 signalling complexes after ligand stimulation. Moreover, cytokine production in Rip2-deficient cells was reduced on stimulation of TLRs with lipopolysaccharide, peptidoglycan and double-stranded RNA, but not with bacterial DNA, indicating that Rip2 is downstream of TLR2/3/4 but not TLR9. Rip2-deficient cells were also hyporesponsive to signalling through interleukin (IL)-1 and IL-18 receptors, and deficient for signalling through Nod proteins-molecules also implicated in the innate immune response. Furthermore, Rip2-deficient T cells showed severely reduced NF-kappaB activation, IL-2 production and proliferation on T-cell-receptor (TCR) engagement, and impaired differentiation to T-helper subtype 1 (TH1) cells, indicating that Rip2 is required for optimal TCR signalling and T-cell differentiation. Rip2 is therefore a signal transducer and integrator of signals for both the innate and adaptive immune systems.


Subject(s)
Adaptor Proteins, Signal Transducing , Drosophila Proteins , Immunity, Innate/immunology , Intracellular Signaling Peptides and Proteins , Listeriosis/immunology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Carrier Proteins/metabolism , Cell Differentiation , Cells, Cultured , Gene Deletion , Interleukins/biosynthesis , Interleukins/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Listeria monocytogenes/immunology , Listeria monocytogenes/physiology , Listeriosis/metabolism , Listeriosis/pathology , Lymphocyte Activation , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein , Nod2 Signaling Adaptor Protein , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cytokine/biosynthesis , Receptors, Interleukin/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptor 2 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...