Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 525
Filtrar
1.
Front Immunol ; 15: 1374293, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38680489

RESUMO

Introduction: Shigella is the etiologic agent of a bacillary dysentery known as shigellosis, which causes millions of infections and thousands of deaths worldwide each year due to Shigella's unique lifestyle within intestinal epithelial cells. Cell adhesion/invasion assays have been extensively used not only to identify targets mediating host-pathogen interaction, but also to evaluate the ability of Shigella-specific antibodies to reduce virulence. However, these assays are time-consuming and labor-intensive and fail to assess differences at the single-cell level. Objectives and methods: Here, we developed a simple, fast and high-content method named visual Adhesion/Invasion Inhibition Assay (vAIA) to measure the ability of anti-Shigellaantibodies to inhibit bacterial adhesion to and invasion of epithelial cells by using the confocal microscope Opera Phenix. Results: We showed that vAIA performed well with a pooled human serum from subjects challenged with S. sonnei and that a specific anti-IpaD monoclonal antibody effectively reduced bacterial virulence in a dose-dependent manner. Discussion: vAIA can therefore inform on the functionality of polyclonal and monoclonal responses thereby supporting the discovery of pathogenicity mechanisms and the development of candidate vaccines and immunotherapies. Lastly, this assay is very versatile and may be easily applied to other Shigella species or serotypes and to different pathogens.


Assuntos
Anticorpos Antibacterianos , Aderência Bacteriana , Disenteria Bacilar , Humanos , Aderência Bacteriana/imunologia , Disenteria Bacilar/imunologia , Disenteria Bacilar/microbiologia , Disenteria Bacilar/diagnóstico , Anticorpos Antibacterianos/imunologia , Interações Hospedeiro-Patógeno/imunologia , Shigella/imunologia , Shigella/patogenicidade , Células Epiteliais/microbiologia , Células Epiteliais/imunologia , Shigella sonnei/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Células HeLa
2.
J Vis Exp ; (204)2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38407235

RESUMO

The human-adapted enteric bacterial pathogen Shigella causes millions of infections each year, creates long-term growth effects among pediatric patients, and is a leading cause of diarrheal deaths worldwide. Infection induces watery or bloody diarrhea as a result of the pathogen transiting the gastrointestinal tract and infecting the epithelial cells lining the colon. With staggering increases in antibiotic resistance and the current lack of approved vaccines, standardized research protocols are critical to studying this formidable pathogen. Here, methodologies are presented to examine the molecular pathogenesis of Shigella using in vitro analyses of bacterial adherence, invasion, and intracellular replication in colonic epithelial cells. Prior to infection analyses, the virulence phenotype of Shigella colonies was verified by the uptake of the Congo red dye on agar plates. Supplemented laboratory media can also be considered during bacterial culturing to mimic in vivo conditions. Bacterial cells are then used in a standardized protocol to infect colonic epithelial cells in tissue culture plates at an established multiplicity of infection with adaptations to analyze each stage of infection. For adherence assays, Shigella cells are incubated with reduced media levels to promote bacterial contact with epithelial cells. For both invasion and intracellular replication assays, gentamicin is applied for various time intervals to eliminate extracellular bacteria and enable assessment of invasion and/or the quantification of intracellular replication rates. All infection protocols enumerate adherent, invaded, and/or intracellular bacteria by serially diluting infected epithelial cell lysates and plating bacterial colony forming units relative to infecting titers on Congo red agar plates. Together, these protocols enable independent characterization and comparisons for each stage of Shigella infection of epithelial cells to study this pathogen successfully.


Assuntos
Disenteria Bacilar , Shigella , Humanos , Criança , Ágar , Vermelho Congo , Células Epiteliais , Diarreia
3.
Eur J Cell Biol ; 103(1): 151381, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38183814

RESUMO

The facultative intracellular pathogen Shigella flexneri invades non-phagocytic epithelial gut cells. Through a syringe-like apparatus called type 3 secretion system, it injects effector proteins into the host cell triggering actin rearrangements leading to its uptake within a tight vacuole, termed the bacterial-containing vacuole (BCV). Simultaneously, Shigella induces the formation of large vesicles around the entry site, which we refer to as infection-associated macropinosomes (IAMs). After entry, Shigella ruptures the BCV and escapes into the host cytosol by disassembling the BCV remnants. Previously, IAM formation has been shown to be required for efficient BCV escape, but the molecular events associated with BCV disassembly have remained unclear. To identify host components required for BCV disassembly, we performed a microscopy-based screen to monitor the recruitment of BAR domain-containing proteins, which are a family of host proteins involved in membrane shaping and sensing (e.g. endocytosis and recycling) during Shigella epithelial cell invasion. We identified endosomal recycling BAR protein Sorting Nexin-8 (SNX8) localized to IAMs in a PI(3)P-dependent manner before BCV disassembly. At least two distinct IAM subpopulations around the BCV were found, either being recycled back to cellular compartments such as the plasma membrane or transitioning to become RAB11A positive "contact-IAMs" involved in promoting BCV rupture. The IAM subpopulation duality was marked by the exclusive recruitment of either SNX8 or RAB11A. Hindering PI(3)P production at the IAMs led to an inhibition of SNX8 recruitment at these compartments and delayed both, the step of BCV rupture time and successful BCV disassembly. Finally, siRNA depletion of SNX8 accelerated BCV rupture and unpeeling of BCV remnants, indicating that SNX8 is involved in controlling the timing of the cytosolic release. Overall, our work sheds light on how Shigella establishes its intracellular niche through the subversion of a specific set of IAMs.


Assuntos
Fosfatos de Fosfatidilinositol , Shigella , Humanos , Shigella/fisiologia , Vacúolos/metabolismo , Células Epiteliais/fisiologia , Shigella flexneri/genética , Células HeLa , Nexinas de Classificação/metabolismo
4.
J Ethnopharmacol ; 324: 117788, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38296176

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Shigella infection is a public health problem responsible for approximately 700,000 deaths annually. The management of this disease is impaired by the emergence of multidrug-resistant Shigella species, highlighting the urgent need to search for alternative treatment options. In this regard, investigating medicinal plants traditionally used for the treatment of dysentery, diarrheal infections, and/or associated symptoms in endemic regions might provide an opportunity to identify phytochemicals that could be further used as a basis for the development of future anti-shigella drug candidates. AIM OF THE STUDY: This study was designed to investigate the anti-shigella and antioxidant-based ethnopharmacological potency of some Cameroonian medicinal plants with an emphasis on pharmacokinetic properties of the identified chemical pharmacophore. MATERIALS AND METHODS: Briefly, plant species were selected and collected based on their ethnopharmacological uses and information reported in the literature. Crude aqueous, ethanolic, methanolic, and hydroethanolic (30:70, v/v) extracts from these plants were prepared and then screened for their anti-Shigella activity against four Shigella strains and cytotoxicity against Vero and Raw cell lines using microdilution and resazurin-based methods, respectively. The antioxidant activities of potent extracts were evaluated using DPPH, ABTS, NO, and FRAP scavenging assays. The chemical profile of potent extracts was performed using the UHPLC-LIT-MS/MS and the pharmacokinetic properties, druglikeness, and likely molecular targets of the chemical scaffolds identified were predicted using SwissADME and SwissTargetPredictor. RESULTS: Thirty-nine (39) plants belonging to 26 plant families were harvested. Out of the 228 extracts tested, 18 extracts originating from 6 plants (15.38 %) were active (MICs 250-1000 µg/mL) and nontoxic toward Vero (CC50 129.25-684.55 µg/mL) and Raw cell lines (CC50 336.20 to >1000 µg/mL). Six potent extracts from the two plants exhibited moderate to potent DPPH (SC50 8.870-54.410 µg/mL), ABTS (SC50 12.020-27.36 µg/mL), and NO (SC50 0.02-195.85 µg/mL) scavenging activities. Later, these extracts showed interesting ferric iron-reducing power (1.28-12.14 µg equivalent NH2OH/g of extract). The shortest onset of action time (4 and 6 h) observed following inhibition kinetics studies was observed with extracts BFSHE, PMSE, and PMSM. The UHPLC-LIT-MS/MS and some databases (Mass Spectral Library (NIST 14), Human Metabolome Database (HMD), MassBank, SuperNatural 3.0, The Food Database (FooDB), and Chemical Entities of Biological Interest (ChEBI)) allowed the annotation of 18 and 17 metabolites in the extracts from stem bark of P. macrophylla and B. ferruginea respectively. Pharmacokinetic prediction of these chemicals showed that compound 6 (4,6a-bis(Hydroxymethyl)-9a-methyl-3-oxo-1a,1b,3,5,6,6a,7a,9a-octahydrobis (oxireno)[2',3':5,6; 2″,3'':9,10]cyclodeca[1,2-b]furan-5-yl methacrylate), compound 8 (Corynoxeine), and compounds 35 (Stachybotrydial acetate) demonstrated acceptable druglike and pharmacokinetic properties and might act through inhibition of kinase, transferase, protease, oxidoreductase, and family AG protein-linked receptors. CONCLUSION: The findings from this investigation demonstrated that Cameroonian medicinal plants are suitable reservoirs of anti-Shigella and antioxidant agents with good drug candidate properties.


Assuntos
Benzotiazóis , Plantas Medicinais , Shigella , Ácidos Sulfônicos , Humanos , Plantas Medicinais/química , Extratos Vegetais/uso terapêutico , Antioxidantes/farmacologia , Antioxidantes/química , Espectrometria de Massas em Tandem , Cromatografia Líquida de Alta Pressão , Camarões
5.
mSphere ; 8(5): e0015423, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37565760

RESUMO

Shigella flexneri is a facultative intracellular pathogen that causes shigellosis, a human diarrheal disease characterized by the destruction of the colonic epithelium. Novel antimicrobial compounds to treat infections are urgently needed due to the proliferation of bacterial antibiotic resistance and lack of new effective antimicrobials in the market. Our approach to find compounds that block the Shigella virulence pathway has three potential advantages: (i) resistance development should be minimized due to the lack of growth selection pressure, (ii) no resistance due to environmental antibiotic exposure should be developed since the virulence pathways are not activated outside of host infection, and (iii) the normal intestinal microbiota, which do not have the targeted virulence pathways, should be unharmed. We chose to utilize two phenotypic assays, inhibition of Shigella survival in macrophages and Shigella growth inhibition (minimum inhibitory concentration), to interrogate the 1.7 M compound screening collection subset of the GlaxoSmithKline drug discovery chemical library. A number of secondary assays on the hit compounds resulting from the primary screens were conducted, which, in combination with chemical, structural, and physical property analyses, narrowed the final hit list to 44 promising compounds for further drug discovery efforts. The rapid development of antibiotic resistance is a critical problem that has the potential of returning the world to a "pre-antibiotic" type of environment, where millions of people will die from previously treatable infections. One relatively newer approach to minimize the selection pressures for the development of resistance is to target virulence pathways. This is anticipated to eliminate any resistance selection pressure in environmental exposure to virulence-targeted antibiotics and will have the added benefit of not affecting the non-virulent microbiome. This paper describes the development and application of a simple, reproducible, and sensitive assay to interrogate an extensive chemical library in high-throughput screening format for activity against the survival of Shigella flexneri 2457T-nl in THP-1 macrophages. The ability to screen very large numbers of compounds in a reasonable time frame (~1.7 M compounds in ~8 months) distinguishes this assay as a powerful tool in further exploring new compounds with intracellular effect on S. flexneri or other pathogens with similar pathways of pathogenesis. The assay utilizes a luciferase reporter which is extremely rapid, simple, relatively inexpensive, and sensitive and possesses a broad linear range. The assay also utilized THP-1 cells that resemble primary monocytes and macrophages in morphology and differentiation properties. THP-1 cells have advantages over human primary monocytes or macrophages because they are highly plastic and their homogeneous genetic background minimizes the degree of variability in the cell phenotype (1). The intracellular and virulence-targeted selectivity of our methodology, determined via secondary screening, is an enormous advantage. Our main interest focuses on hits that are targeting virulence, and the most promising compounds with adequate physicochemical and drug metabolism and pharmacokinetic (DMPK) properties will be progressed to a suitable in vivo shigellosis model to evaluate the therapeutic potential of this approach. Additionally, compounds that act via a host-directed mechanism could be a promising source for further research given that it would allow a whole new, specific, and controlled approach to the treatment of diseases caused by some pathogenic bacteria.


Assuntos
Disenteria Bacilar , Shigella , Humanos , Shigella flexneri , Virulência/genética , Disenteria Bacilar/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Macrófagos
6.
Semin Immunol ; 70: 101812, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37562110

RESUMO

Besides its crucial function in nutrient absorbance and as barrier against the microbiota, the gut epithelium is essential for sensing pathogenic insults and mounting of an appropriate early immune response. In mice, the activation of the canonical NAIP/NLRC4 inflammasome is critical for the defense against enterobacterial infections. Activation of the NAIP/NLRC4 inflammasome triggers the extrusion of infected intestinal epithelial cells (IEC) into the gut lumen, concomitant with inflammasome-mediated lytic cell death. The membrane permeabilization, a hallmark of pyroptosis, is caused by the pore-forming proteins called gasdermins (GSDMs). Recent work has revealed that NAIP/NLRC4-dependent extrusion of infected IECs can, however, also be executed in the absence of GSDMD. In fact, several reports highlighted that various cell death pathways (e.g., pyroptosis or apoptosis) and unique mechanisms specific to particular infection models and stages of gut infection are in action during epithelial inflammasome defense against intestinal pathogens. Here, we summarize the current knowledge regarding the underlying mechanisms and speculate on the putative functions of the epithelial inflammasome activation and cell death, with a particular emphasis on mouse infection models for two prominent enterobacterial pathogens, Salmonella Typhimurium and Shigella flexneri.


Assuntos
Inflamassomos , Shigella , Camundongos , Animais , Humanos , Gasderminas , Salmonella typhimurium/metabolismo , Shigella/metabolismo , Inflamação
7.
Front Cell Infect Microbiol ; 13: 1183211, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37389216

RESUMO

Shigella is a Gram-negative bacterial pathogen that relies on a single type three secretion system (T3SS) as its primary virulence factor. The T3SS includes a highly conserved needle-like apparatus that directly injects bacterial effector proteins into host cells, subverting host cell function, initiating infection, and circumventing resulting host immune responses. Recent findings have located the T3SS ATPase Spa47 to the base of the Shigella T3SS apparatus and have correlated its catalytic function to apparatus formation, protein effector secretion, and overall pathogen virulence. This critical correlation makes Spa47 ATPase activity regulation a likely point of native control over Shigella virulence and a high interest target for non-antibiotic- based therapeutics. Here, we provide a detailed characterization of the natural 11.6 kDa C-terminal translation product of the Shigella T3SS protein Spa33 (Spa33C), showing that it is required for proper virulence and that it pulls down with several known T3SS proteins, consistent with a structural role within the sorting platform of the T3SS apparatus. In vitro binding assays and detailed kinetic analyses suggest an additional role, however, as Spa33C differentially regulates Spa47 ATPase activity based on Spa47s oligomeric state, downregulating Spa47 monomer activity and upregulating activity of both homo-oligomeric Spa47 and the hetero-oligomeric MxiN2Spa47 complex. These findings identify Spa33C as only the second known differential T3SS ATPase regulator to date, with the Shigella protein MxiN representing the other. Describing this differential regulatory protein pair begins to close an important gap in understanding of how Shigella may modulate virulence through Spa47 activity and T3SS function.


Assuntos
Adenosina Trifosfatases , Shigella , Proteínas de Bactérias/genética , Catálise , Movimento Celular
8.
Nature ; 616(7957): 590-597, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36991122

RESUMO

Gasdermins (GSDMs) are pore-forming proteins that play critical roles in host defence through pyroptosis1,2. Among GSDMs, GSDMB is unique owing to its distinct lipid-binding profile and a lack of consensus on its pyroptotic potential3-7. Recently, GSDMB was shown to exhibit direct bactericidal activity through its pore-forming activity4. Shigella, an intracellular, human-adapted enteropathogen, evades this GSDMB-mediated host defence by secreting IpaH7.8, a virulence effector that triggers ubiquitination-dependent proteasomal degradation of GSDMB4. Here, we report the cryogenic electron microscopy structures of human GSDMB in complex with Shigella IpaH7.8 and the GSDMB pore. The structure of the GSDMB-IpaH7.8 complex identifies a motif of three negatively charged residues in GSDMB as the structural determinant recognized by IpaH7.8. Human, but not mouse, GSDMD contains this conserved motif, explaining the species specificity of IpaH7.8. The GSDMB pore structure shows the alternative splicing-regulated interdomain linker in GSDMB as a regulator of GSDMB pore formation. GSDMB isoforms with a canonical interdomain linker exhibit normal pyroptotic activity whereas other isoforms exhibit attenuated or no pyroptotic activity. Overall, this work sheds light on the molecular mechanisms of Shigella IpaH7.8 recognition and targeting of GSDMs and shows a structural determinant in GSDMB critical for its pyroptotic activity.


Assuntos
Proteínas de Bactérias , Gasderminas , Proteínas Citotóxicas Formadoras de Poros , Animais , Humanos , Camundongos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Sítios de Ligação , Sequência Conservada , Microscopia Crioeletrônica , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Citotóxicas Formadoras de Poros/ultraestrutura , Domínios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/ultraestrutura , Piroptose , Shigella , Especificidade da Espécie , Gasderminas/química , Gasderminas/metabolismo , Gasderminas/ultraestrutura
9.
J Appl Microbiol ; 134(2)2023 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-36626757

RESUMO

AIMS: A rapid rise in resistance to conventional antibiotics for Shigella spp. has created a problem in treating shigellosis. Hence, there is an urgent need for new and non-conventional anti-bacterial agents. The aim of this study is to show how Asiatic acid, a plant-derived compound, inhibits the intracellular growth of Shigella flexneri. METHODS AND RESULTS: Shigella flexneri sensitive and resistant strains were used for checking antimicrobial activity of Asiatic acid by gentamicin protection assay. Asiatic acid inhibited the intracellular growth of all strains. Gene expression analysis showed antimicrobial peptide (AMP) up-regulation by Asiatic acid in intestinal cells. Further western blot analysis showed that ERK, p38, and JNK are activated by Asiatic acid. ELISA was performed to check IL-8, IL-6, and cathelicidin secretion. The antibacterial effect of Asiatic acid was further verified in an in vivo mouse model. CONCLUSIONS: The reason behind the antibacterial activities of Asiatic acid is probably over-expression of antimicrobial peptide genes. Besides, direct antimicrobial activities, antimicrobial peptides also carry immunomodulatory activities. Here, Asiatic acid increased IL-6 and IL-8 secretion to induce inflammation. Overall, Asiatic acid up-regulates antimicrobial peptide gene expression and inhibits intracellular S. flexneri growth. Moreover, Asiatic acid reduced bacterial growth and recovered intestinal tissue damages in in vivo mice model.


Assuntos
Disenteria Bacilar , Shigella , Animais , Camundongos , Antibacterianos/farmacologia , Disenteria Bacilar/tratamento farmacológico , Disenteria Bacilar/microbiologia , Expressão Gênica , Interleucina-6/genética , Interleucina-8/genética , Testes de Sensibilidade Microbiana , Shigella/genética , Shigella flexneri/genética , Peptídeos Antimicrobianos/farmacologia
10.
Am J Trop Med Hyg ; 108(1): 174-180, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36509064

RESUMO

Diarrhea is a common cause of morbidity and mortality among children younger than 5 years in developing countries. Children from 3 to 60 months of age were recruited from two hospitals in Nepal- Bharatpur Hospital, Bharatpur, and Kanti Children's Hospital, Kathmandu-in 2006 to 2009. Stool specimens collected from 1,200 children with acute diarrhea (cases) and 1,200 children without diarrhea (control subjects) were examined for a broad range of enteropathogens by standard microbiology, including microscopy, enzyme immunoassay for viral pathogens (adenovirus, astrovirus, and rotavirus) and protozoa (Giardia, Cryptosporidium, and Entamoeba histolytica), as well as by using reverse transcription real-time polymerase for norovirus. Antimicrobial susceptibility testing was performed using the disk diffusion method. Overall, rotavirus (22% versus 2%), norovirus (13% versus 7%), adenovirus (3% versus 0%), Shigella (6% versus 1%), enterotoxigenic Escherichia coli (8% versus 4%), Vibrio (7% versus 0%), and Aeromonas (9% versus 3%) were identified significantly more frequently in cases than control subjects. Campylobacter, Plesiomonas, Salmonella, and diarrheagenic E. coli (enteropathogenic, enteroinvasive, enteroaggregative) were identified in similar proportions in diarrheal and non-diarrheal stools. Campylobacter was resistant to second-generation quinolone drugs (ciprofloxacin and norfloxacin), whereas Vibrio and Shigella were resistant to nalidixic acid and trimethoprim/sulfamethoxazole. This study documents the important role of rotavirus and norovirus in acute diarrhea in children younger than 5 years, followed by the bacteria Shigella, enterotoxigenic E. coli, Vibrio cholera, and Aeromonas. Data on the prevalence and epidemiology of enteropathogens identify potential pathogens for public health interventions, whereas pathogen antibiotic resistance pattern data may provide guidance on choice of therapy in clinical settings.


Assuntos
Infecções por Adenoviridae , Anti-Infecciosos , Campylobacter , Criptosporidiose , Cryptosporidium , Escherichia coli Enterotoxigênica , Norovirus , Rotavirus , Shigella , Humanos , Lactente , Pré-Escolar , Nepal/epidemiologia , Diarreia/microbiologia , Adenoviridae , Doença Aguda
11.
Microbiol Spectr ; 10(6): e0341022, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36374106

RESUMO

Shigella IcsA is a versatile surface virulence factor required for early and late pathogenesis stages extracellularly and intracellularly. Despite IcsA serving as a model Type V secretion system (T5SS) autotransporter to study host-pathogen interactions, its detailed molecular architecture is poorly understood. Recently, IcsA was found to switch to a different conformation for its adhesin activity upon sensing the host stimuli by Shigella Type III secretion system (T3SS). Here, we reported that the single cysteine residue (C130) near the N terminus of the IcsA passenger had a role in IcsA adhesin activity. We also showed that the IcsA passenger (IcsAp) existed in multiple conformations, and the conformation populations were influenced by a central pair of cysteine residues (C375 and C379), which was not previously reported for any Type V autotransporter passengers. Disruption of either or both central cysteine residues altered the exposure of IcsA epitopes to polyclonal anti-IcsA antibodies previously shown to block Shigella adherence, yet without loss of IcsA intracellular functions in actin-based motility (ABM). Anti-IcsA antibody reactivity was restored when the IcsA-paired cysteine substitution mutants were expressed in an ΔipaD background with a constitutively active T3SS, highlighting an interplay between T3SS and T5SS. The work here uncovered a novel molecular switch empowered by a centrally localized, short-spaced cysteine pair in the Type V autotransporter IcsA that ensured conformational heterogeneity to aid IcsA evasion of host immunity. IMPORTANCE Shigella species are the leading cause of diarrheal-related death globally by causing bacillary dysentery. The surface virulence factor IcsA, which is essential for Shigella pathogenesis, is a unique multifunctional autotransporter that is responsible for cell adhesion, and actin-based motility, yet detailed mechanistic understanding is lacking. Here, we showed that the three cysteine residues in IcsA contributed to the protein's distinct functions. The N-terminal cysteine residue within the IcsA passenger domain played a role in adhesin function, while a centrally localized cysteine pair provided conformational heterogeneity that resulted in IcsA molecules with different reactivity to adhesion-blocking anti-IcsA antibodies. In synergy with the Type III secretion system, this molecular switch preserved biological function in distinct IcsA conformations for cell adhesion, actin-based motility, and autophagy escape, providing a potential strategy by which Shigella evades host immunity and targets this essential virulence factor.


Assuntos
Proteínas de Ligação a DNA , Shigella , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Shigella flexneri/genética , Sistemas de Secreção Tipo V/genética , Sistemas de Secreção Tipo V/metabolismo , Cisteína/metabolismo , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Actinas/metabolismo , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
12.
Microbiol Spectr ; 10(6): e0192622, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36350161

RESUMO

Tuberculous meningitis (TBM), the most lethal and disabling form of tuberculosis (TB), may be related to gut microbiota composition, warranting further study. Here we systematically compared gut microbiota compositions and blood cytokine profiles of TBM patients, pulmonary TB patients, and healthy controls. Notably, the significant gut microbiota dysbiosis observed in TBM patients was associated with markedly high proportions of Escherichia-Shigella species as well as increased blood levels of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6). Next, we obtained a fecal bacterial isolate from a TBM patient and administered it via oral gavage to mice in order to develop a murine gut microbiota dysbiosis model for use in exploring mechanisms underlying the observed relationship between gut microbial dysbiosis and TBM. Thereafter, cells of commensal Escherichia coli (E. coli) were isolated and administered to model mice by gavage and then mice were inoculated with Mycobacterium tuberculosis (M. tuberculosis). Subsequently, these mice exhibited increased blood TNF-α levels accompanied by downregulated expression of tight junction protein claudin-5, increased brain tissue bacterial burden, and elevated central nervous system inflammation relative to corresponding indicators in controls administered PBS by gavage. Thus, our results demonstrated that a signature dysbiotic gut microbiome profile containing a high proportion of E. coli was potentially associated with an increased circulating TNF-α level in TBM patients. Collectively, these results suggest that modulation of dysbiotic gut microbiota holds promise as a new strategy for preventing or alleviating TBM. IMPORTANCE As the most severe form of tuberculosis, the pathogenesis of tuberculous meningitis (TBM) is still unclear. Gut microbiota dysbiosis plays an important role in a variety of central nervous system diseases. However, the relationship between gut microbiota and TBM has not been identified. In our study, significant dysbiosis in gut microbiota composition with a high proportion of E. coli and increased levels of TNF-α in plasma was noted in TBM patients. A commensal E. coli was isolated and shown to increase the plasma level of TNF-α and downregulate brain tight junction protein claudin-5 in the murine model. Gavage administration of E. coli aggravated the bacterial burden and increased the inflammatory responses in the central nervous system after M. tuberculosis infection. Dysbiosis of gut microbiota may be a promising therapeutic target and biomarker for TBM prevention or treatment.


Assuntos
Microbioma Gastrointestinal , Mycobacterium tuberculosis , Shigella , Tuberculose Meníngea , Camundongos , Animais , Fator de Necrose Tumoral alfa/metabolismo , Microbioma Gastrointestinal/fisiologia , Escherichia coli/metabolismo , Disbiose/microbiologia , Claudina-5 , Mycobacterium tuberculosis/metabolismo
13.
Front Cell Infect Microbiol ; 12: 1012533, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36389142

RESUMO

Shigella, the causative agent of bacillary dysentery, subvert cytoskeletal and trafficking processes to invade and replicate in epithelial cells using an arsenal of bacterial effectors translocated through a type III secretion system. Here, we review the various roles of the type III effector IpgD, initially characterized as phosphatidylinositol 4,5 bisphosphate (PI4,5P2) 4-phosphatase. By decreasing PI4,5P2 levels, IpgD triggers the disassembly of cortical actin filaments required for bacterial invasion and cell migration. PI5P produced by IpgD further stimulates signaling pathways regulating cell survival, macropinosome formation, endosomal trafficking and dampening of immune responses. Recently, IpgD was also found to exhibit phosphotransferase activity leading to PI3,4P2 synthesis adding a new flavor to this multipotent bacterial enzyme. The substrate of IpgD, PI4,5P2 is also the main substrate hydrolyzed by endogenous phospholipases C to produce inositoltriphosphate (InsP3), a major Ca2+ second messenger. Hence, beyond the repertoire of effects associated with the direct diversion of phoshoinositides, IpgD indirectly down-regulates InsP3-mediated Ca2+ release by limiting InsP3 production. Furthermore, IpgD controls the intracellular lifestyle of Shigella promoting Rab8/11 -dependent recruitment of the exocyst at macropinosomes to remove damaged vacuolar membrane remnants and promote bacterial cytosolic escape. IpgD thus emerges as a key bacterial effector for the remodeling of host cell membranes.


Assuntos
Disenteria Bacilar , Shigella , Humanos , Fosfatidilinositóis/metabolismo , Shigella flexneri/metabolismo , Disenteria Bacilar/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo
14.
Curr Opin Cell Biol ; 79: 102131, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36215855

RESUMO

Subversion of the host cell cytoskeleton is a virulence attribute common to many bacterial pathogens. On mucosal surfaces, bacteria have evolved distinct ways of interacting with the polarised epithelium and manipulating host cell structure to propagate infection. For example, Shigella and Listeria induce cytoskeletal changes to induce their own uptake into enterocytes in order to replicate within an intracellular environment and then spread from cell-to-cell by harnessing the host actin cytoskeleton. In this review, we highlight some recent studies that advance our understanding of the role of the host cell cytoskeleton in the mechanical and molecular processes of pathogen invasion, cell-to-cell spread and the impact of infection on epithelial intercellular tension and innate mucosal defence.


Assuntos
Listeria , Shigella , Citoesqueleto/metabolismo , Células Epiteliais , Bactérias , Interações Hospedeiro-Patógeno
15.
Microb Drug Resist ; 28(11): 1043-1055, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36130139

RESUMO

Aims: This study represents the first analysis from Iran for both the frequency of the most common causes of infectious diarrhoea and their antibiotic resistance patterns in adult patients. Methods: Adult stool specimens (n = 211) were analyzed. Stool specimens were analyzed using standard microbiological, polymerase chain reaction, and reverse transcription polymerase chain reaction tests to identify bacterial, parasitic, and viral enteropathogens. Antibiotic resistance profiles were determined. Results: Enteropathogens were identified in 46.4% (98/211) of the surveyed samples. This included 33.1% (70/211) bacterial infections, including 9.9% (21/211) diarrheagenic Escherichia coli (DEC) and 8.5% (18/211) Shigella spp. We detected 7.1% (15/211) parasitic infections (mostly Giardia lamblia) and 6.1% (13/211) viral infections (mostly adenovirus). The DEC and Shigella spp. isolates included many multi-drug resistant (MDR) isolates (95.2% and 77.7%, respectively), and extended spectrum-ß-lactamase (ESBL) genes were often present (57.1% and 61.1%, respectively). The most commonly identified ESBL genes in the DEC and Shigella spp. isolates were blaTEM (100% in both species), blaCTX-M15 (91.6% and 100%, respectively), AmpC blaCIT (80% and 100%, respectively), and blaDHA (80% and 100%, respectively). Conclusions: Bacterial infection was the primary cause of infectious diarrhea, affecting one-third of the adults. The frequency of DEC and Shigella spp. was higher than for other enteropathogens. The high prevalence of MDR, the elevated incidence of ESBL genes among Shigella spp. and DEC isolates, and the presence of quinolone resistance in the Salmonella spp. isolates represent a significant challenge for gastroenteritis diagnosis and treatment in this region.


Assuntos
Infecções por Escherichia coli , Gastroenterite , Shigella , Humanos , Adulto , beta-Lactamases/genética , Testes de Sensibilidade Microbiana , Irã (Geográfico)/epidemiologia , Antibacterianos/farmacologia , Shigella/genética , Gastroenterite/tratamento farmacológico , Gastroenterite/epidemiologia , Infecções por Escherichia coli/microbiologia
16.
Comput Biol Med ; 148: 105900, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35952542

RESUMO

Shigella is a Gram-negative bacteria that cause shigellosis. Treatment with antibiotics cannot be sustained to control the bacterial infection due to the risk of antibiotic resistance. Vaccine development against the highly prevalent Shigella serotypes could provide a generous benefit in reducing the occurrence of shigellosis. The present study is aimed to identify the peptides that could be the ideal candidates for the Shigella vaccine development. THP-1 human macrophage cell lines were infected with clinical strains of Shigella flexneri 2a. The bacterial peptides bound on HLA class II molecules of infected THP-1 were analyzed and identified using the immunopeptidomics approach. Following mass spectrometry identification, a total of 14 proteins were predicted by PSORTb, CELLO, and Gneg-mPLoc as outer membrane proteins (OMPs) of Shigella. Of which, 12 OMPs were found to be conserved among Shigella species and had no significance with human proteomes. Outer membrane receptor FepA and TonB-dependent receptor were among the OMPs predicted to possess the high number of immunogenic B- and T-cell epitopes. The epitopes with high antigenicity from FepA and TonB were identified as potential peptide candidates for Shigella vaccine development. The immunoreactivity of the constructed recombinant proteins were determined using the Shigella-infected human and rabbit sera, respectively. Their protective efficacy and immune responses in controlling the Shigella infection will further be investigated in experimental animal models.


Assuntos
Disenteria Bacilar , Shigella , Animais , Epitopos de Linfócito T , Humanos , Espectrometria de Massas , Peptídeos , Coelhos , Vacinologia
17.
mBio ; 13(4): e0053822, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35924851

RESUMO

Enteropathogenic Escherichia coli (EPEC) and Shigella are etiologic agents of diarrhea in children <5 years old living in resource-poor countries. Repeated bouts of infection lead to lifelong morbidity and even death. The goal of this study was to characterize local mucosal immune responses in Shigella- and EPEC-infected children <5 years of age with moderate to severe diarrhea (MSD) enrolled in the Global Enteric Multicenter Study (GEMS). We hypothesized that infection with each of these pathogens would induce distinct gut mucosal immune profiles indicative of disease etiology and severity. To test this hypothesis, innate and adaptive immune markers were measured in stools from children with diarrhea due to EPEC, Shigella, or other organisms and in children who had no diarrhea. Shigella-positive diarrhea evoked robust proinflammatory and TH1/TH2 cytokine responses compared to diarrhea caused by EPEC or other organisms, with the exception of interleukin 5 (IL-5), which was associated with EPEC infection. The presence of IL-1ß, IL-4, IL-16, and tumor necrosis factor beta (TNF-ß) was associated with the absence of dysentery. EPEC-positive diarrhea evoked high levels of IL-1ß, vascular endothelial growth factor (VEGF), and IL-10. Granulocyte-macrophage colony-stimulating factor (GM-CSF) had opposing roles in disease severity, being associated with absence of diarrhea in EPEC-infected children and with dysenteric Shigella infection. High levels of antigen-specific antibodies were detected in the controls and children with Shigella without dysentery, which suggests a protective role against severe disease. In summary, this study identified distinct local immune responses associated with two clinically relevant diarrheagenic pathogens, Shigella and EPEC, in children and identified protective immune phenotypes that can inform the development of preventive measures. IMPORTANCE Shigella and enteropathogenic Escherichia coli are primary agents of moderate to severe diarrhea in children <5 years of age living in resource-poor countries. Repeated bouts of illness lead to lifelong health impairment and even death. Aiming to understand the local host immunity to these pathogens in relation to disease prognosis and to identify prophylaxis and therapeutic targets, we investigated innate and adaptive immune profiles in stools from children infected with EPEC with and without diarrhea, Shigella with and without dysentery, and controls in well characterized clinical samples obtained during the Global Enteric Multicenter Study. For the first time, we report pathogen-specific mucosal immune profiles associated with severity or absence of disease in children <5 years of age that can inform prevention and treatment efforts.


Assuntos
Disenteria , Escherichia coli Enteropatogênica , Infecções por Escherichia coli , Shigella , Diarreia , Disenteria/complicações , Infecções por Escherichia coli/complicações , Humanos , Índice de Gravidade de Doença , Shigella/genética , Fator A de Crescimento do Endotélio Vascular
18.
mBio ; 13(3): e0094422, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35652591

RESUMO

Polymorphonuclear neutrophils (PMN) are recruited to the gastrointestinal mucosa in response to inflammation, injury, and infection. Here, we report the development and the characterization of an ex vivo tissue coculture model consisting of human primary intestinal enteroid monolayers and PMN, and a mechanistic interrogation of PMN-epithelial cell interaction and response to Shigella, a primary cause of childhood dysentery. Cellular adaptation and tissue integration, barrier function, PMN phenotypic and functional attributes, and innate immune responses were examined. PMN within the enteroid monolayers acquired a distinct activated/migratory phenotype that was influenced by direct epithelial cell contact as well as by molecular signals. Seeded on the basal side of the intestinal monolayer, PMN were intercalated within the epithelial cells and moved paracellularly toward the apical side. Cocultured PMN also increased basal secretion of interleukin 8 (IL-8). Shigella added to the apical surface of the monolayers evoked additional PMN phenotypic adaptations, including increased expression of cell surface markers associated with chemotaxis and cell degranulation (CD47, CD66b, and CD88). Apical Shigella infection triggered rapid transmigration of PMN to the luminal side, neutrophil extracellular trap (NET) formation, and bacterial phagocytosis and killing. Shigella infection modulated cytokine production in the coculture; apical monocyte chemoattractant protein (MCP-1), tumor necrosis factor alpha (TNF-α), and basolateral IL-8 production were downregulated, while basolateral IL-6 secretion was increased. We demonstrated, for the first time, PMN phenotypic adaptation and mobilization and coordinated epithelial cell-PMN innate response upon Shigella infection in the human intestinal environment. The enteroid monolayer-PMN coculture represents a technical innovation for mechanistic interrogation of gastrointestinal physiology, host-microbe interaction, innate immunity, and evaluation of preventive/therapeutic tools. IMPORTANCE Studies of mucosal immunity and microbial host cell interaction have traditionally relied on animal models and in vitro tissue culture using immortalized cancer cell lines, which yield nonphysiological and often unreliable results. Herein, we report the development and characterization of an ex vivo enteroid-PMN coculture consisting of normal human intestinal epithelium and a mechanistic interrogation of PMN and epithelial cell interaction and function in the context of Shigella infection. We demonstrated tissue-driven phenotypic and functional adaptation of PMN and a coordinated epithelial cell and PMN response to Shigella, a primary cause of dysentery in young children in the developing world.


Assuntos
Disenteria Bacilar , Shigella , Animais , Células Cultivadas , Pré-Escolar , Técnicas de Cocultura , Disenteria Bacilar/metabolismo , Células Epiteliais/metabolismo , Humanos , Interleucina-8 , Mucosa Intestinal/metabolismo , Neutrófilos , Shigella/metabolismo
19.
Front Immunol ; 13: 894206, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35769459

RESUMO

Helicobacter pylori (H. pylori) is a gram-negative pathogen classified as a class I carcinogen. The H. pylori urease B subunit (UreB) and heat shock protein A (HspA) are two important vaccine candidate antigens. In this study, we evaluated the immunogenicity and immunoprotective effect of the attenuated Shigella vector vaccine SH02 expressing the UreB-HspA fusion protein of H. pylori in a mouse model. Oral SH02 with or without subcutaneous injection of rUreB-HspA induced antigen-specific serum IgG, mucosal sIgA, and T cells immune response. Subcutaneous injection of the candidate antigen rUreB-HspA enhanced the level of serum antigen-specific IgG antibodies (p < 0.0001) and the levels of IgG1/IgG2a/IgG2b subtypes. In addition, injection boost also increased the proportion of spleen antigen-specific CD4+CD154+ T cells (p < 0.001), and the proportion of CD4+CD154+ T cells that secrete IFN-γ and IL-17A. Following the H. pylori challenge, the levels of H. pylori colonization in the two experimental groups (Groups A and B) significantly reduced compared with the control group (p < 0.001), indicating that the candidate vaccine yielded a preventive effect of anti-H.pylori infection. Compared with the non-subcutaneous booster injection group (Group A), the subcutaneous booster injection group (Group B) exhibited less gastric inflammation, but there was no significant difference in the level of colonization (p > 0.05). These results lay a foundation for the development of a vaccine against H. pylori and the optimization of immunization methods and procedures to prevent H. pylori infection.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Vacinas contra Shigella , Shigella , Administração Oral , Animais , Antígenos de Bactérias/genética , Vacinas Bacterianas , Modelos Animais de Doenças , Infecções por Helicobacter/prevenção & controle , Imunoglobulina G , Camundongos , Camundongos Endogâmicos BALB C , Proteína Estafilocócica A , Vacinas Atenuadas
20.
Vaccine ; 40(30): 3991-3998, 2022 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-35660036

RESUMO

Shigella is a well-known etiological agent responsible for intestinal infection among children, the elderly, and immunocompromised people ranging from mild to severe cases. Shigellosis remains endemic in Malaysia and yet there is no commercial vaccine available to eradicate the disease. Iron is an essential element for the survival of Shigella within the host. Hence, it is required for regulating metabolic mechanisms and virulence determinants. Alteration of iron status in the extracellular environment directly triggers the signal in enteropathogenic bacterial, providing information that they are in a hostile environment. To survive in an iron-limited environment, molecular regulation of iron-binding proteins plays a vital role in facilitating the transportation and utilization of sufficient iron sources. Given the importance of iron molecules for bacterial survival and pathogenicity, this review summarizes the physiological role of iron-binding proteins in bacterial survival and their potential use in vaccine and therapeutic developments.


Assuntos
Disenteria Bacilar , Shigella , Idoso , Proteínas de Bactérias/metabolismo , Criança , Humanos , Ferro/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Desenvolvimento de Vacinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA