Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 26(5): 106738, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37216124

RESUMEN

A crypt autochthonous microbial population called crypt-associated microbiota (CAM) is localized intimately with gut regenerative and immune machinery. The present report utilizes laser capture microdissection coupled with 16S amplicon sequencing to characterize the CAM in patients with ulcerative colitis (UC) before and after fecal microbiota transplantation with anti-inflammatory diet (FMT-AID). Compositional differences in CAM and its interactions with mucosa-associated microbiota (MAM) were compared between the non-IBD controls and in patients with UC pre- and post-FMT (n = 26). Distinct from the MAM, CAM is dominated by aerobic members of Actinobacteria and Proteobacteria and exhibits resilience of diversity. CAM underwent UC-associated dysbiosis and demonstrated restoration post-FMT-AID. These FMT-restored CAM taxa correlated negatively with disease activity in patients with UC. The positive effects of FMT-AID extended further in refurbishing CAM-MAM interactions, which were obliterated in UC. These results encourage investigation into host-microbiome interactions established by CAM, to understand their role in disease pathophysiology.

2.
Nanoscale ; 14(39): 14717-14731, 2022 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-36169577

RESUMEN

Proinflammatory cytokines such as Tumor Necrosis Factor-α (TNF-α) are critical mediators of inflammatory bowel disease pathogenesis, and are important targets to restore intestinal homeostasis. Herein, we present the engineering and screening of gemini lipid nanoparticles (GLNPs) for siRNA delivery to colon epithelial cells, macrophages and dendritic cells, and their ability to deliver siRNA therapeutics to the inflamed gastrointestinal tract. We synthesized eight gemini cationic lipids by tethering two lithocholic acid molecules through 3'-hydroxyl- and 24'-carboxyl-derived ammonium groups using different polyalkylene spacers. Screening of GLNPs, composed of gemini cationic lipid and dioleoylphosphatidylethanolamine lipid, showed that GLNPs derived from gemini lipid G1 are the most effective in the delivery of siRNA across mammalian cell membranes with reduced toxicity. Gemini lipid G1-derived siRNA-GLNP complexes (siGLNPs) can effectively reduce gene expression, and are stable in simulated gastric fluid. The delivery of TNF-α siRNA using siGLNPs can mitigate gut inflammation in a dextran sodium sulfate-induced murine inflammation model. As CD4+ T cells, especially Th17 cells, are key mediators of gut inflammation, we further showed that these siGLNPs inhibit infiltration and differentiation of CD4+ T cells to Th17 and Treg cells. Therefore, this study highlights the potential of GLNPs derived from lithocholic acid-derived gemini cationic lipids for the development of next-generation nucleic acid delivery vehicles.


Asunto(s)
Compuestos de Amonio , Factor de Necrosis Tumoral alfa , Animales , Linfocitos T CD4-Positivos , Cationes , Citocinas , Dextranos , Inflamación , Lípidos , Liposomas , Ácido Litocólico , Mamíferos/genética , Ratones , Nanopartículas , ARN Interferente Pequeño
3.
Gut Microbes ; 13(1): 1986665, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34696686

RESUMEN

Non-typhoidal Salmonella (NTS) infections result in self limiting gastroenteritis except in rare cases wherein manifestations of chronic infections can occur. Strategies employed by Salmonella to thrive in hostile environments of host during chronic infections are complex and multifaceted. In chronic state, a coordinated action of bacterial effectors allows reprogramming of macrophages to M2 subtype and thereby creating a permissible replicative niche. The mechanistic details of these processes are not fully known. In the current study we identified, histone H3-lysine 27 trimethylation (H3K27me3)-specific demethylase, KDM6B to be upregulated in both cell culture and in murine model of Salmonella infection. KDM6B recruitment upon infection exhibited an associated loss of overall H3K27me3 in host cells and was Salmonella SPI1 effectors coordinated. ChIP-qRT-PCR array analysis revealed several new gene promoter targets of KDM6B demethylase activity including PPARδ, a crucial regulator of fatty acid oxidation pathway and Salmonella-persistent infections. Furthermore, pharmacological inhibition of KDM6B demethylase activity with GSKJ4 in chronic Salmonella infection mice model led to a significant reduction in pathogen load and M2 macrophage polarization in peripheral lymphoid organs. The following work thus reveals Salmonella effector-mediated epigenetic reprogramming of macrophages responsible for its long-term survival and chronic carriage.


Asunto(s)
Epigénesis Genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Infecciones por Salmonella/enzimología , Salmonella typhimurium/fisiología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Histonas/genética , Histonas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Metilación , Ratones , Ratones Endogámicos C57BL , PPAR delta/genética , PPAR delta/metabolismo , Infecciones por Salmonella/genética , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/genética , Regulación hacia Arriba
4.
Mol Aspects Med ; 81: 100997, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34311996

RESUMEN

The genus Salmonella represents a wide range of strains including Typhoidal and Non-Typhoidal Salmonella (NTS) isolates that exhibit illnesses of varied pathophysiologies. The more frequent NTS ensues a self-limiting enterocolitis with rare occasions of bacteremia or systemic infections. These self-limiting Salmonella strains are capable of subverting and dampening the host immune system to achieve a more prolonged survival inside the host system thus leading to chronic manifestations. Notably, emergence of new invasive NTS isolates known as invasive Non-Typhoidal Salmonella (iNTS) have worsened the disease burden significantly in some parts of the world. NTS strains adapt to attain persister phenotype intracellularly and cause relapsing infections. These chronic infections, in susceptible hosts, are also capable of causing diseases like IBS, IBD, reactive arthritis, gallbladder cancer and colorectal cancer. The present understanding of molecular mechanism of how these chronic infections are manifested is quite limited. The current work is an effort to review the prevailing knowledge emanating from a large volume of research focusing on various forms of NTS infections including those that cause localized, systemic and persistent disease. The review will further dwell into the understanding of how this pathogen contributes to the associated long term sequelae.


Asunto(s)
Bacteriemia , Infecciones por Salmonella , Humanos , Infección Persistente , Fenotipo , Salmonella/genética
5.
Biomater Sci ; 9(5): 1481-1502, 2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33404019

RESUMEN

Inflammatory bowel disease (IBD) is an autoimmune disorder of the gastrointestinal tract (GIT) where Ulcerative Colitis (UC) displays localized inflammation in the colon, and Crohn's Disease (CD) affects the entire GIT. Failure of current therapies and associated side-effects bring forth serious social, economic, and health challenges. The gut epithelium provides the best target for gene therapy delivery vehicles to combat IBD. Gene therapy involving the use of nucleic acid (NA) therapeutics faces major challenges due to the hydrophilic, negative-charge, and degradable nature of NAs. Recent success in the engineering of biomaterials for gene therapy and their emergence in clinical trials for various diseases is an inspiration for scientists to develop gene therapy vehicles that can be easily targeted to the desired tissues for IBD. Advances in nanotechnology have enabled the formulations of numerous nanoparticles for NA delivery to mitigate IBD that still faces challenges of stability in the GIT, poor therapeutic efficacy, and targetability. This review presents the challenges of gene therapeutics, gastrointestinal barriers, and recent advances in the engineering of nanoparticles for IBD treatment along with future directions for successful translation of nanoparticle-mediated gene therapeutics in clinics.


Asunto(s)
Colitis Ulcerosa , Colitis , Enfermedades Inflamatorias del Intestino , Nanopartículas , Terapia Genética , Humanos , Enfermedades Inflamatorias del Intestino/terapia
7.
Nanoscale ; 11(11): 4970-4986, 2019 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-30839018

RESUMEN

Poor success rates and challenges associated with the current therapeutic strategies of inflammatory bowel disease (IBD) have accelerated the emergence of gene therapy as an alternative treatment option with great promise. However, oral delivery of nucleic acids (NAs) to an inflamed colon is challenged by multiple barriers presented by the gastrointestinal, extracellular and intracellular compartments. Therefore, we screened a series of polyaspartic acid-derived amphiphilic cationic polymers with varied hydrophobicity for their ability to deliver NAs into mammalian cells. Using the most effective TAC6 polymer, we then engineered biocompatible and stable nanogels composed of polyplexes (TAC6, NA) and an anionic polymer, sodium polyaspartate, that were able to deliver the NAs across mammalian cells using caveolae-mediated cellular uptake. We then utilized these nanogels for oral delivery of PIAS1 (protein inhibitor of activated STAT1), a SUMO 3 ligase, encoding plasmid DNA since PIAS1 is a key nodal therapeutic target for IBD due to its ability to control NF-κB-mediated inflammatory signaling. We show that plasmid delivery using TAC6-derived nanogels diminished gut inflammation in a murine colitis model. Therefore, our study presents engineering of orally deliverable nanogels that can target SUMOylation machinery to combat gut inflammation with very high efficacy.


Asunto(s)
Colitis/terapia , Técnicas de Transferencia de Gen/instrumentación , Terapia Genética/métodos , Polietilenglicoles/administración & dosificación , Polietileneimina/administración & dosificación , Sumoilación , Administración Oral , Animales , Cationes/química , Línea Celular Tumoral , Colitis/patología , Colitis/fisiopatología , Colon/metabolismo , Colon/patología , Colon/fisiopatología , Modelos Animales de Enfermedad , Endocitosis , Expresión Génica , Terapia Genética/instrumentación , Humanos , Inflamación , Ratones , Nanogeles , Péptidos/química , Plásmidos/administración & dosificación , Plásmidos/química , Plásmidos/genética , Polietilenglicoles/química , Polietilenglicoles/metabolismo , Polietileneimina/química , Polietileneimina/metabolismo , Polímeros/química , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Inhibidoras de STAT Activados/metabolismo
8.
J Cell Sci ; 132(1)2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30510112

RESUMEN

Salmonella Typhimurium is an intracellular pathogen that causes gastroenteritis in humans. Aided by a battery of effector proteins, S. Typhimurium resides intracellularly in a specialized vesicle, called the Salmonella-containing vacuole (SCV) that utilizes the host endocytic vesicular transport pathway (VTP). Here, we probed the possible role of SUMOylation, a post-translation modification pathway, in SCV biology. Proteome analysis by complex mass-spectrometry (MS/MS) revealed a dramatically altered SUMO-proteome (SUMOylome) in S. Typhimurium-infected cells. RAB7, a component of VTP, was key among several crucial proteins identified in our study. Detailed MS/MS assays, in vitro SUMOylation assays and structural docking analysis revealed SUMOylation of RAB7 (RAB7A) specifically at lysine 175. A SUMOylation-deficient RAB7 mutant (RAB7K175R) displayed longer half-life, was beneficial to SCV dynamics and functionally deficient. Collectively, the data revealed that RAB7 SUMOylation blockade by S. Typhimurium ensures availability of long-lived but functionally compromised RAB7, which was beneficial to the pathogen. Overall, this SUMOylation-dependent switch of RAB7 controlled by S. Typhimurium is an unexpected mode of VTP pathway regulation, and unveils a mechanism of broad interest well beyond Salmonella-host crosstalk. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Vesículas Citoplasmáticas/patología , Células Epiteliales/microbiología , Mucosa Intestinal/microbiología , Infecciones por Salmonella/patología , Salmonella typhimurium/patogenicidad , Sumoilación , Proteínas de Unión al GTP rab/metabolismo , Células Cultivadas , Vesículas Citoplasmáticas/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Interacciones Huésped-Patógeno , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/crecimiento & desarrollo , Proteínas de Unión al GTP rab/química , Proteínas de Unión a GTP rab7
9.
Open Biol ; 7(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28659381

RESUMEN

Post-translational modification pathways such as SUMOylation are integral to all cellular processes and tissue homeostasis. We investigated the possible involvement of SUMOylation in the epithelial signalling in Crohn's disease (CD) and ulcerative colitis (UC), the two major forms of inflammatory bowel disease (IBD). Initially in a murine model of IBD, induced by dextran-sulfate-sodium (DSS mice), we observed inflammation accompanied by a lowering of global SUMOylation of colonic epithelium. The observed SUMOylation alteration was due to a decrease in the sole SUMO E2 enzyme (Ubc9). Mass-spectrometric analysis revealed the existence of a distinct SUMOylome (SUMO-conjugated proteome) in DSS mice with alteration of key cellular regulators, including master kinase Akt1. Knocking-down of Ubc9 in epithelial cells resulted in dramatic activation of inflammatory gene expression, a phenomenon that acted via reduction in Akt1 and its SUMOylated form. Importantly, a strong decrease in Ubc9 and Akt1 was also seen in endoscopic biopsy samples (N = 66) of human CD and UC patients. Furthermore, patients with maximum disease indices were always accompanied by severely lowered Ubc9 or SUMOylated-Akt1. Mucosal tissues with severely compromised Ubc9 function displayed higher levels of pro-inflammatory cytokines and compromised wound-healing markers. Thus, our results reveal an important and previously undescribed role for the SUMOylation pathway involving Ubc9 and Akt1 in modulation of epithelial inflammatory signalling in IBD.


Asunto(s)
Regulación hacia Abajo , Enfermedades Inflamatorias del Intestino/fisiopatología , Mucosa Intestinal/enzimología , Sumoilación , Enzimas Activadoras de Ubiquitina/genética , Enzimas Activadoras de Ubiquitina/metabolismo , Adolescente , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Persona de Mediana Edad , Transducción de Señal/genética , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Adulto Joven
10.
Mol Cell Biol ; 35(17): 2932-46, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26100020

RESUMEN

Posttranslational modifications (PTMs) can alter many fundamental properties of a protein. One or combinations of them have been known to regulate the dynamics of many cellular pathways and consequently regulate all vital processes. Understandably, pathogens have evolved sophisticated strategies to subvert these mechanisms to achieve instantaneous control over host functions. Here, we present the first report of modulation by intestinal pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) of host SUMOylation, a PTM pathway central to all fundamental cellular processes. Both in cell culture and in a mouse model, we observed that S. Typhimurium infection led to a dynamic SUMO-conjugated proteome alteration. The intracellular survival of S. Typhimurium was dependent on SUMO status as revealed by reduced infection and Salmonella-induced filaments (SIFs) in SUMO-upregulated cells. S. Typhimurium-dependent SUMO modulation was seen as a result of depletion of crucial SUMO pathway enzymes Ubc-9 and PIAS1, at both the protein and the transcript levels. Mechanistically, depletion of Ubc-9 relied on upregulation of small noncoding RNAs miR30c and miR30e during S. Typhimurium infection. This was necessary and sufficient for both down-modulation of Ubc-9 and a successful infection. Thus, we demonstrate a novel strategy of pathogen-mediated perturbation of host SUMOylation, an integral mechanism underlying S. Typhimurium infection and intracellular survival.


Asunto(s)
MicroARNs/genética , Proteínas Inhibidoras de STAT Activados/metabolismo , Salmonella typhimurium/patogenicidad , Sumoilación/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Animales , Línea Celular Tumoral , Femenino , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/patología
11.
Cell Microbiol ; 17(6): 843-59, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25486861

RESUMEN

Salmonella enterica Typhimurium induces intestinal inflammation through the activity of type III secreted effector (T3SE) proteins. Our prior results indicate that the secretion of the T3SE SipA and the ability of SipA to induce epithelial cell responses that lead to induction of polymorphonuclear transepithelial migration are not coupled to its direct delivery into epithelial cells from Salmonella. We therefore tested the hypothesis that SipA interacts with a membrane protein located at the apical surface of intestinal epithelial cells. Employing a split ubiquitin yeast-two-hybrid screen, we identified the tetraspanning membrane protein, p53 effector related to PMP-22 (PERP), as a SipA binding partner. SipA and PERP appear to have intersecting activities as we found PERP to be involved in proinflammatory pathways shown to be regulated by SipA. In sum, our studies reveal a critical role for PERP in the pathogenesis of S. Typhimurium, and for the first time demonstrate that SipA, a T3SE protein, can engage a host protein at the epithelial surface.


Asunto(s)
Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Inflamación/microbiología , Inflamación/patología , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Salmonella typhimurium/inmunología , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Genes Supresores de Tumor , Humanos , Unión Proteica , Mapeo de Interacción de Proteínas , Migración Transendotelial y Transepitelial , Técnicas del Sistema de Dos Híbridos
12.
Cell Mol Life Sci ; 68(22): 3687-97, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21984608

RESUMEN

Acute gastroenteritis caused by Salmonella enterica serovar typhimurium is a significant public health problem. This pathogen has very sophisticated molecular machinery encoded by the two pathogenicity islands, namely Salmonella Pathogenicity Island 1 and 2 (SPI-1 and SPI-2). Remarkably, both SPI-1 and SPI-2 are very tightly regulated in terms of timing of expression and spatial localization of the encoded effectors during the infection process within the host cell. This regulation is governed at several levels, including transcription and translation, and by post-translational modifications. In the context of a finely tuned regulatory system, we will highlight how these effector proteins co-opt host signaling pathways that control the ability of the organism to infect and survive within the host, as well as elicit host pro-inflammatory responses.


Asunto(s)
Proteínas Bacterianas/inmunología , Interacciones Huésped-Patógeno/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Animales , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Islas Genómicas , Humanos , Inmunidad Innata/inmunología , Procesamiento Proteico-Postraduccional , Infecciones por Salmonella/fisiopatología , Salmonella typhimurium/genética , Transducción de Señal/fisiología , Vacuolas/metabolismo , Vacuolas/microbiología
13.
Science ; 330(6002): 390-393, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20947770

RESUMEN

The enteric pathogen Salmonella enterica serovar Typhimurium causes food poisoning resulting in gastroenteritis. The S. Typhimurium effector Salmonella invasion protein A (SipA) promotes gastroenteritis by functional motifs that trigger either mechanisms of inflammation or bacterial entry. During infection of intestinal epithelial cells, SipA was found to be responsible for the early activation of caspase-3, an enzyme that is required for SipA cleavage at a specific recognition motif that divided the protein into its two functional domains and activated SipA in a manner necessary for pathogenicity. Other caspase-3 cleavage sites identified in S. Typhimurium appeared to be restricted to secreted effector proteins, which indicates that this may be a general strategy used by this pathogen for processing of its secreted effectors.


Asunto(s)
Proteínas Bacterianas/metabolismo , Caspasa 3/metabolismo , Mucosa Intestinal/microbiología , Proteínas de Microfilamentos/metabolismo , Salmonelosis Animal/microbiología , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Línea Celular Tumoral , Activación Enzimática , Gastroenteritis/metabolismo , Gastroenteritis/microbiología , Gastroenteritis/patología , Humanos , Mucosa Intestinal/enzimología , Intestinos/enzimología , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Infiltración Neutrófila , Salmonelosis Animal/patología , Factores de Virulencia/metabolismo
14.
Oncotarget ; 1(8): 721-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21321381

RESUMEN

When one considers the organism Salmonella enterica serotype Typhimurium (S. Typhimurium), one usually thinks of the Gram-negative enteric pathogen that causes the severe food borne illness, gastroentertitis. In this context, the idea of Salmonella being exploited as a cancer therapeutic seems pretty remote. However, there has been an escalating interest in the development of tumor-therapeutic bacteria for use in the treatment of a variety of cancers. This strategy takes advantage of the remarkable ability of certain bacteria to preferentially replicate and accumulate within tumors. In the case of S. Typhimurium, this organism infects and selectively grows within implanted tumors, achieving tumor/normal tissue ratios of approximately 1,000:1. Salmonella also has some attractive properties well suited for the design of a chemotherapeutic agent. In particular, this pathogen can easily be manipulated to carry foreign genes, and since this species is a facultative anaerobe, it is able to survival in both oxygenated and hypoxic conditions, implying this organism could colonize both small metastatic lesions as well as larger tumors. These observations are the impetus to a burgeoning field focused on the development of Salmonella as a clinically useful anti-cancer agent. We will discuss three cutting edge technologies employing Salmonella to target tumors.


Asunto(s)
Toxinas Bacterianas/uso terapéutico , Terapia Molecular Dirigida/métodos , Neoplasias/terapia , Salmonella typhimurium/fisiología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Humanos , Modelos Biológicos , Salmonella typhimurium/metabolismo
15.
J Bacteriol ; 191(21): 6525-38, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19717599

RESUMEN

The distinctive feature of the GroES-GroEL chaperonin system in mediating protein folding lies in its ability to exist in a tetradecameric state, form a central cavity, and encapsulate the substrate via the GroES lid. However, recombinant GroELs of Mycobacterium tuberculosis are unable to act as effective molecular chaperones when expressed in Escherichia coli. We demonstrate here that the inability of M. tuberculosis GroEL1 to act as a functional chaperone in E. coli can be alleviated by facilitated oligomerization. The results of directed evolution involving random DNA shuffling of the genes encoding M. tuberculosis GroEL homologues followed by selection for functional entities suggested that the loss of chaperoning ability of the recombinant mycobacterial GroEL1 and GroEL2 in E. coli might be due to their inability to form canonical tetradecamers. This was confirmed by the results of domain-swapping experiments that generated M. tuberculosis-E. coli chimeras bearing mutually exchanged equatorial domains, which revealed that E. coli GroEL loses its chaperonin activity due to alteration of its oligomerization capabilities and vice versa for M. tuberculosis GroEL1. Furthermore, studying the oligomerization status of native GroEL1 from cell lysates of M. tuberculosis revealed that it exists in multiple oligomeric forms, including single-ring and double-ring variants. Immunochemical and mass spectrometric studies of the native M. tuberculosis GroEL1 revealed that the tetradecameric form is phosphorylated on serine-393, while the heptameric form is not, indicating that the switch between the single- and double-ring variants is mediated by phosphorylation.


Asunto(s)
Chaperonina 60/metabolismo , Mycobacterium tuberculosis/metabolismo , Alelos , Secuencia de Aminoácidos , Chaperonina 60/genética , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Prueba de Complementación Genética , Modelos Moleculares , Chaperonas Moleculares , Mycobacterium tuberculosis/genética , Fosforilación
16.
Interdiscip Perspect Infect Dis ; 2008: 626827, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19259328

RESUMEN

The mucosal surfaces of the gastrointestinal tract harbor a vast number of commensal microbiota that have coevolved with the host, and in addition display one of the most complex relationships with the host. This relationship affects several important aspects of the biology of the host including the synthesis of nutrients, protection against infection, and the development of the immune system. On the other hand, despite the existence of several lines of mucosal defense mechanisms, pathogenic organisms such as Shigella and Salmonella have evolved sophisticated virulence strategies for breaching these barriers. The constant challenge from these pathogens and the attempts by the host to counter them set up a dynamic equilibrium of cellular and molecular crosstalk. Even slight perturbations in this equilibrium may be detrimental to the host leading to severe bacterial infection or even autoimmune diseases like inflammatory bowel disease. Several experimental model systems, including germ-free mice and antibiotic-treated mice, have been used by various researchers to study this complex relationship. Although it is only the beginning, it promises to be an exciting era in the study of these host-microbe relationships.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...