Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Gut Microbes ; 15(2): 2265138, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37842920

RESUMO

Recently, an intestinal dysbiotic microbiota with enrichment in oral cavity bacteria has been described in colorectal cancer (CRC) patients. Here, we characterize and investigate one of these oral pathobionts, the Gram-positive anaerobic coccus Parvimonas micra. We identified two phylotypes (A and B) exhibiting different phenotypes and adhesion capabilities. We observed a strong association of phylotype A with CRC, with its higher abundance in feces and in tumoral tissue compared with the normal homologous colonic mucosa, which was associated with a distinct methylation status of patients. By developing an in vitro hypoxic co-culture system of human primary colonic cells with anaerobic bacteria, we show that P. micra phylotype A alters the DNA methylation profile promoters of key tumor-suppressor genes, oncogenes, and genes involved in epithelial-mesenchymal transition. In colonic mucosa of CRC patients carrying P. micra phylotype A, we found similar DNA methylation alterations, together with significant enrichment of differentially expressed genes in pathways involved in inflammation, cell adhesion, and regulation of actin cytoskeleton, providing evidence of P. micra's possible role in the carcinogenic process.


Assuntos
Neoplasias Colorretais , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/genética , Firmicutes/genética , Bactérias , Neoplasias Colorretais/genética , Neoplasias Colorretais/microbiologia
3.
Sci Rep ; 13(1): 14960, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37696912

RESUMO

In this work, we investigated the oncogenic role of Streptococcus gallolyticus subsp. gallolyticus (SGG), a gut bacterium associated with colorectal cancer (CRC). We showed that SGG UCN34 accelerates colon tumor development in a chemically induced CRC murine model. Full proteome and phosphoproteome analysis of murine colons chronically colonized by SGG UCN34 revealed that 164 proteins and 725 phosphorylation sites were differentially regulated. Ingenuity Pathway Analysis (IPA) indicates a pro-tumoral shift specifically induced by SGG UCN34, as ~ 90% of proteins and phosphoproteins identified were associated with digestive cancer. Comprehensive analysis of the altered phosphoproteins using ROMA software revealed up-regulation of several cancer hallmark pathways such as MAPK, mTOR and integrin/ILK/actin, affecting epithelial and stromal colonic cells. Importantly, an independent analysis of protein arrays of human colon tumors colonized with SGG showed up-regulation of PI3K/Akt/mTOR and MAPK pathways, providing clinical relevance to our findings. To test SGG's capacity to induce pre-cancerous transformation of the murine colonic epithelium, we grew ex vivo organoids which revealed unusual structures with compact morphology. Taken together, our results demonstrate the oncogenic role of SGG UCN34 in a murine model of CRC associated with activation of multiple cancer-related signaling pathways.


Assuntos
Neoplasias do Colo , Streptococcus gallolyticus subspecies gallolyticus , Humanos , Animais , Camundongos , Modelos Animais de Doenças , Fosfatidilinositol 3-Quinases , Proteômica , Serina-Treonina Quinases TOR , Fosfoproteínas , Proteoma , Transdução de Sinais
4.
Cell Stem Cell ; 29(5): 856-868.e5, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35523143

RESUMO

After birth, the intestine undergoes major changes to shift from an immature proliferative state to a functional intestinal barrier. By combining inducible lineage tracing and transcriptomics in mouse models, we identify a prodifferentiation PDGFRαHigh intestinal stromal lineage originating from postnatal LTßR+ perivascular stromal progenitors. The genetic blockage of this lineage increased the intestinal stem cell pool while decreasing epithelial and immune maturation at weaning age, leading to reduced postnatal growth and dysregulated repair responses. Ablating PDGFRα in the LTBR stromal lineage demonstrates that PDGFRα has a major impact on the lineage fate and function, inducing a transcriptomic switch from prostemness genes, such as Rspo3 and Grem1, to prodifferentiation factors, including BMPs, retinoic acid, and laminins, and on spatial organization within the crypt-villus and repair responses. Our results show that the PDGFRα-induced transcriptomic switch in intestinal stromal cells is required in the first weeks after birth to coordinate postnatal intestinal maturation and function.


Assuntos
Intestinos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Animais , Diferenciação Celular/fisiologia , Mecanismos de Defesa , Mucosa Intestinal , Receptor beta de Linfotoxina , Camundongos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Células-Tronco
5.
EMBO Rep ; 23(2): e54341, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-34914162

RESUMO

SARS-CoV-2 infection results in impaired interferon response in patients with severe COVID-19. However, how SARS-CoV-2 interferes with host immune responses is incompletely understood. Here, we sequence small RNAs from SARS-CoV-2-infected human cells and identify a microRNA (miRNA) derived from a recently evolved region of the viral genome. We show that the virus-derived miRNA produces two miRNA isoforms in infected cells by the enzyme Dicer, which are loaded into Argonaute proteins. Moreover, the predominant miRNA isoform targets the 3'UTR of interferon-stimulated genes and represses their expression in a miRNA-like fashion. Finally, the two viral miRNA isoforms were detected in nasopharyngeal swabs from COVID-19 patients. We propose that SARS-CoV-2 can potentially employ a virus-derived miRNA to hijack the host miRNA machinery, which could help to evade the interferon-mediated immune response.


Assuntos
COVID-19 , MicroRNAs , RNA Viral/genética , SARS-CoV-2/genética , Regiões 3' não Traduzidas , COVID-19/imunologia , Humanos , Imunidade , MicroRNAs/genética
6.
J Vis Exp ; (175)2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34605823

RESUMO

Reactive oxygen species (ROS) play essential roles in intestinal homeostasis. ROS are natural by-products of cell metabolism. They are produced in response to infection or injury at the mucosal level as they are involved in antimicrobial responses and wound healing. They are also critical secondary messengers, regulating several pathways, including cell growth and differentiation. On the other hand, excessive ROS levels lead to oxidative stress, which can be deleterious for cells and favor intestinal diseases like chronic inflammation or cancer. This work provides a straightforward method to detect ROS in the intestinal murine organoids by live imaging and flow cytometry, using a commercially available fluorogenic probe. Here the protocol describes assaying the effect of compounds that modulate the redox balance in intestinal organoids and detect ROS levels in specific intestinal cell types, exemplified here by the analysis of the intestinal stem cells genetically labeled with GFP. This protocol may be used with other fluorescent probes.


Assuntos
Organoides , Estresse Oxidativo , Animais , Intestinos , Camundongos , Oxirredução , Espécies Reativas de Oxigênio
7.
Cell Host Microbe ; 27(3): 358-375.e7, 2020 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-32101704

RESUMO

Despite the recognized capacity of the gut microbiota to regulate intestinal lipid metabolism, the role of specific commensal species remains undefined. Here, we aimed to understand the bacterial effectors and molecular mechanisms by which Lactobacillus paracasei and Escherichia coli regulate lipid metabolism in enterocytes. We show that L-lactate produced by L. paracasei inhibits chylomicron secretion from enterocytes and promotes lipid storage by a mechanism involving L-lactate absorption by enterocytes, its conversion to malonyl-CoA, and the subsequent inhibition of lipid beta-oxidation. In contrast, acetate produced by E. coli also inhibits chylomicron secretion by enterocytes but promotes lipid oxidation by a mechanism involving acetate absorption by enterocytes, its metabolism to acetyl-CoA and AMP, and the subsequent upregulation of the AMPK/PGC-1α/PPARα pathway. Our study opens perspectives for developing specific bacteria- and metabolite-based therapeutic interventions against obesity, atherosclerosis, and malnutrition by targeting lipid metabolism in enterocytes.


Assuntos
Enterócitos/metabolismo , Escherichia coli/metabolismo , Fermentação , Lacticaseibacillus paracasei/metabolismo , Metabolismo dos Lipídeos , Simbiose , Animais , Linhagem Celular , Quilomícrons , Enterócitos/microbiologia , Feminino , Intestinos/microbiologia , Camundongos Endogâmicos C57BL
8.
Proc Natl Acad Sci U S A ; 117(4): 1994-2003, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31919280

RESUMO

The nucleotide-binding oligomerization domain-containing protein 2 (NOD2) agonist muramyl dipeptide (MDP), a peptidoglycan motif common to all bacteria, supports leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5)+ intestinal stem cell (ISC) survival through NOD2 activation upon an otherwise lethal oxidative stress-mediated signal. However, the underlying protective mechanisms remain unknown. Here, using irradiation as stressor and primarily murine-derived intestinal organoids as a model system, we show that MDP induced a significant reduction of total and mitochondrial reactive oxygen species (ROS) within ISCs, which was associated with mitophagy induction. ATG16L1 knockout (KO) and NOD2 KO organoids did not benefit from the MDP-induced cytoprotection. We confirmed the MDP-dependent induction of ISC mitophagy upon stress in vivo. These findings elucidate the NOD2-mediated mechanism of cytoprotection involving the clearance of the lethal excess of ROS molecules through mitophagy, triggered by the coordinated activation of NOD2 and ATG16L1 by a nuclear factor κB (NF-κB)-independent pathway.


Assuntos
Acetilmuramil-Alanil-Isoglutamina/farmacologia , Citoproteção , Intestinos/crescimento & desenvolvimento , Mitofagia/efeitos dos fármacos , Proteína Adaptadora de Sinalização NOD2/metabolismo , Espécies Reativas de Oxigênio , Células-Tronco/citologia , Animais , Células Cultivadas , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Imunidade Inata/efeitos da radiação , Intestinos/efeitos dos fármacos , Intestinos/efeitos da radiação , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mitofagia/efeitos da radiação , Proteína Adaptadora de Sinalização NOD2/genética , Estresse Oxidativo/efeitos dos fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/efeitos da radiação
9.
Nat Microbiol ; 5(1): 34-39, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31819216

RESUMO

The gut commensal segmented filamentous bacterium (SFB) attaches to the ileal epithelium and potently stimulates the host immune system. Using transmission electron microscopy (TEM), we show that mouse and rat SFB are flagellated above the concave tip at the unicellular intracellular offspring (IO) stage and that flagellation occurs prior to full IO differentiation and release of IOs from SFB filaments. This finding adds a missing link to the SFB life cycle.


Assuntos
Bactérias Anaeróbias/crescimento & desenvolvimento , Bactérias Anaeróbias/ultraestrutura , Flagelos/ultraestrutura , Animais , Linhagem Celular , Flagelos/metabolismo , Flagelina/genética , Flagelina/metabolismo , Regulação Bacteriana da Expressão Gênica , Humanos , Íleo/microbiologia , Mucosa Intestinal/microbiologia , Camundongos , Ratos , Receptor 5 Toll-Like/metabolismo
10.
Pathog Dis ; 77(7)2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31578543

RESUMO

Shigella spp. are bacterial pathogens that invade the human colonic mucosa using a type III secretion apparatus (T3SA), a proteinaceous device activated upon contact with host cells. Active T3SAs translocate proteins that carve the intracellular niche of Shigella spp. Nevertheless, the activation state of the T3SA has not been addressed in vivo. Here, we used a green fluorescent protein transcription-based secretion activity reporter (TSAR) to provide a spatio-temporal description of S. flexneri T3SAs activity in the colon of Guinea pigs. First, we observed that early mucus release is triggered in the vicinity of luminal bacteria with inactive T3SA. Subsequent mucosal invasion showed bacteria with active T3SA associated with the brush border, eventually penetrating into epithelial cells. From 2 to 8 h post-challenge, the infection foci expanded, and these intracellular bacteria displayed homogeneously high-secreting activity, while extracellular foci within the lamina propria featured bacteria with low secretion activity. We also found evidence that within lamina propria macrophages, bacteria reside in vacuoles instead of accessing the cytosol. Finally, bacteria were cleared from tissues between 8 and 24 h post-challenge, highlighting the hit-and-run colonization strategy of Shigella. This study demonstrates how genetically encoded reporters can contribute to deciphering pathogenesis in vivo.


Assuntos
Colo/microbiologia , Disenteria Bacilar/microbiologia , Shigella flexneri/fisiologia , Sistemas de Secreção Tipo III/fisiologia , Animais , Biomarcadores , Modelos Animais de Doenças , Feminino , Genes Reporter , Cobaias , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Especificidade de Órgãos , Distribuição Tecidual
11.
Cell Microbiol ; 21(11): e13118, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31634976

RESUMO

Over the past 10 years, the "Pathogénie Microbienne Moléculaire" unit of Professor Philippe Sansonetti has studied the molecular cross talk between the intestinal microbiota and the gut epithelium, aiming to better understand how this mutualistic symbiosis delineates homoeostasis and, when perturbed, prompts pathology. To do so, the unit has manipulated both bacterial and epithelial cells, and used cutting-edge technology. More recently, the lab has turned its focus also on studying the intestinal crypt and more specifically the intestinal stem cell for their role in epithelial regeneration and long-term epithelium renewal. Here, we provide a brief review summarising recent results obtained from the lab, with particular focus on the intestinal crypt.


Assuntos
Células Epiteliais/microbiologia , Microbioma Gastrointestinal/fisiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Animais , Bactérias/imunologia , Bactérias/metabolismo , Bactérias/patogenicidade , Proliferação de Células , Células Epiteliais/fisiologia , Microbioma Gastrointestinal/imunologia , Homeostase , Interações entre Hospedeiro e Microrganismos , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Regeneração , Células-Tronco/citologia , Células-Tronco/metabolismo , Simbiose
12.
Nat Microbiol ; 4(11): 2001-2009, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31383999

RESUMO

Pathogenic enterobacteria face various oxygen (O2) levels during intestinal colonization from the O2-deprived lumen to oxygenated tissues. Using Shigella flexneri as a model, we have previously demonstrated that epithelium invasion is promoted by O2 in a type III secretion system-dependent manner. However, subsequent pathogen adaptation to tissue oxygenation modulation remained unknown. Assessing single-cell distribution, together with tissue oxygenation, we demonstrate here that the colonic mucosa O2 is actively depleted by S. flexneri aerobic respiration-and not host neutrophils-during infection, leading to the formation of hypoxic foci of infection. This process is promoted by type III secretion system inactivation in infected tissues, favouring colonizers over explorers. We identify the molecular mechanisms supporting infectious hypoxia induction, and demonstrate here how enteropathogens optimize their colonization capacity in relation to their ability to manipulate tissue oxygenation during infection.


Assuntos
Disenteria Bacilar/metabolismo , Mucosa Intestinal/microbiologia , Oxigênio/metabolismo , Shigella flexneri/patogenicidade , Animais , Hipóxia Celular , Modelos Animais de Doenças , Disenteria Bacilar/microbiologia , Feminino , Cobaias , Células Hep G2 , Humanos , Mucosa Intestinal/metabolismo , Coelhos , Shigella flexneri/metabolismo , Sistemas de Secreção Tipo III/metabolismo
13.
Proc Natl Acad Sci U S A ; 116(27): 13582-13591, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31209035

RESUMO

Intracellular trafficking pathways in eukaryotic cells are essential to maintain organelle identity and structure, and to regulate cell communication with its environment. Shigella flexneri invades and subverts the human colonic epithelium by the injection of virulence factors through a type 3 secretion system (T3SS). In this work, we report the multiple effects of two S. flexneri effectors, IpaJ and VirA, which target small GTPases of the Arf and Rab families, consequently inhibiting several intracellular trafficking pathways. IpaJ and VirA induce large-scale impairment of host protein secretion and block the recycling of surface receptors. Moreover, these two effectors decrease clathrin-dependent and -independent endocytosis. Therefore, S. flexneri infection induces a global blockage of host cell intracellular transport, affecting the exchange between cells and their external environment. The combined action of these effectors disorganizes the epithelial cell polarity, disturbs epithelial barrier integrity, promotes multiple invasion events, and enhances the pathogen capacity to penetrate into the colonic tissue in vivo.


Assuntos
Disenteria Bacilar/fisiopatologia , Mucosa Intestinal/microbiologia , Shigella flexneri , Transporte Biológico , Células CACO-2 , Polaridade Celular , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Colo/fisiopatologia , Disenteria Bacilar/metabolismo , Disenteria Bacilar/patologia , Endocitose , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiologia
14.
J Oncol ; 2019: 5415761, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32082377

RESUMO

Helicobacter pylori infection causes chronic gastritis and is the major risk factor of gastric cancer. H. pylori induces a chronic inflammation-producing reactive oxygen species (ROS) which is a source of chromosome instabilities and contributes to the development of malignancy. H. pylori also promotes DNA hypermethylation, known to dysregulate essential genes that maintain genetic stability. The maintenance of telomere length by telomerase is essential for chromosome integrity. Telomerase reverse transcriptase (TERT) is the catalytic component of telomerase activity and an important target during host-pathogen interaction. We aimed to investigate the consequences of H. pylori on the regulation of TERT gene expression and telomerase activity. In vitro, hTERT mRNA levels and telomerase activity were analysed in H. pylori-infected human gastric epithelial cells. In addition, C57BL/6 and INS-GAS mice were used to investigate the influence of H. pylori-induced inflammation on TERT levels. Our data demonstrated that, in vitro, H. pylori inhibits TERT gene expression and decreases the telomerase activity. The exposure of cells to lycopene, an antioxidant compound, restores TERT levels in infected cells, indicating that ROS are implicated in this downregulation. In vivo, fewer TERT-positive cells are observed in gastric tissues of infected mice compared to uninfected, more predominantly in the vicinity of large aggregates of lymphocytes, suggesting an inflammation-mediated regulation. Furthermore, H. pylori appears to downregulate TERT gene expression through DNA hypermethylation as shown by the restoration of TERT transcript levels in cells treated with 5'-azacytidine, an inhibitor of DNA methylation. This was confirmed in infected mice, by PCR-methylation assay of the TERT gene promoter. Our data unraveled a novel way for H. pylori to promote genome instabilities through the inhibition of TERT levels and telomerase activity. This mechanism could play an important role in the early steps of gastric carcinogenesis.

15.
Methods Mol Biol ; 1576: 183-194, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-27628134

RESUMO

The gut, particularly the colon, is the host of approximately 1000 bacterial species, the so-called gut microbiota. The relationship between the gut microbiota and the host is symbiotic and mutualistic, influencing many aspects of the biology of the host. This homeostatic balance can be disrupted by enteric pathogens, such as Shigella flexneri or Listeria monocytogenes, which are able to invade the epithelial layer and consequently subvert physiological functions. To study the host-microbe interactions in vitro, the crypt culture model, known as intestinal organoids, is a powerful tool. Intestinal organoids provide a model in which to examine the response of the epithelium, particularly the response of intestinal stem cells, to the presence of bacteria. Furthermore, the organoid model enables the study of pathogens during the early steps of enteric pathogen invasion.Here, we describe methods that we have established to study the cellular microbiology of symbiosis between the gut microbiota and host intestinal surface and secondly the disruption of host homeostasis due to an enteric pathogen.


Assuntos
Bactérias/crescimento & desenvolvimento , Técnicas de Cultura de Células/métodos , Microbioma Gastrointestinal , Interações Hospedeiro-Patógeno , Mucosa Intestinal/citologia , Organoides/citologia , Células-Tronco/citologia , Diferenciação Celular , Células Cultivadas , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Organoides/metabolismo , Organoides/microbiologia , Células-Tronco/metabolismo , Células-Tronco/microbiologia
16.
EMBO J ; 37(23)2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30389666

RESUMO

While mucosal inflammation is a major source of stress during enteropathogen infection, it remains to be fully elucidated how the host benefits from this environment to clear the pathogen. Here, we show that host stress induced by different stimuli mimicking inflammatory conditions strongly reduces the binding of Shigella flexneri to epithelial cells. Mechanistically, stress activates acid sphingomyelinase leading to host membrane remodeling. Consequently, knockdown or pharmacological inhibition of the acid sphingomyelinase blunts the stress-dependent inhibition of Shigella binding to host cells. Interestingly, stress caused by intracellular Shigella replication also results in remodeling of the host cell membrane, in vitro and in vivo, which precludes re-infection by this and other non-motile pathogens. In contrast, Salmonella Typhimurium overcomes the shortage of permissive entry sites by gathering effectively at the remaining platforms through its flagellar motility. Overall, our findings reveal host membrane remodeling as a novel stress-responsive cell-autonomous defense mechanism that protects epithelial cells from infection by non-motile bacterial pathogens.


Assuntos
Membrana Celular/imunologia , Disenteria Bacilar/imunologia , Células Epiteliais/imunologia , Imunidade Inata , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Shigella flexneri/imunologia , Estresse Fisiológico/imunologia , Animais , Membrana Celular/patologia , Disenteria Bacilar/patologia , Células Epiteliais/patologia , Cobaias , Infecções por Salmonella/patologia
17.
mBio ; 9(5)2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30181250

RESUMO

The gut microbiota contributes to nutrients absorption and metabolism by enterocytes, but the molecular mechanisms involved remain poorly understood, and most conclusions are inferred from studies comparing germfree and conventional animals colonized with diverse bacterial species. We selected two model commensal microorganisms, Escherichia coli and Lactobacillus paracasei, to assess the role of the small-intestinal microbiota in modulating lipid absorption and metabolism by the epithelium. Using an integrated approach encompassing cellular and murine models and combining metabolic parameters measurement, lipid droplet imaging, and gene expression analysis, we demonstrated that under homeostatic conditions, L. paracasei promotes fat storage in enterocytes, whereas E. coli enhances lipid catabolism and reduces chylomicron circulating levels. The Akt/mammalian target of sirolimus (mTOR) pathway is inhibited by both bacterial species in vitro, indicating that several regulatory pathways are involved in the distinct intracellular lipid outcomes associated with each bacterial species. Moreover, soluble bacterial factors partially reproduce the effects observed with live microorganisms. However, reduction of chylomicron circulating levels in E. coli-colonized animals is lost under high-fat-diet conditions, whereas it is potentiated by L. paracasei colonization accompanied by resistance to hypercholesterolemia and excess body weight gain.IMPORTANCE The specific contribution of each bacterial species within a complex microbiota to the regulation of host lipid metabolism remains largely unknown. Using two model commensal microorganisms, L. paracasei and E. coli, we demonstrated that both bacterial species impacted host lipid metabolism in a diet-dependent manner and, notably, that L. paracasei-colonized mice but not E. coli-colonized mice resisted high-fat-diet-induced body weight gain. In addition, we set up cellular models of fatty acid absorption and secretion by enterocytes cocultured with bacteria and showed that, in vitro, both L. paracasei and E. coli inhibited lipid secretion, through increased intracellular fat storage and enhanced lipid catabolism, respectively.


Assuntos
Enterócitos/metabolismo , Escherichia coli/fisiologia , Interações entre Hospedeiro e Microrganismos , Lacticaseibacillus paracasei/fisiologia , Metabolismo dos Lipídeos , Animais , Quilomícrons/sangue , Dieta Hiperlipídica , Feminino , Microbioma Gastrointestinal , Lipídeos/biossíntese , Redes e Vias Metabólicas , Camundongos , Camundongos Endogâmicos C57BL , Simbiose , Serina-Treonina Quinases TOR/fisiologia , Aumento de Peso
18.
Cell Chem Biol ; 25(4): 483-493.e9, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29478905

RESUMO

Neutrophils represent the most abundant immune cells recruited to inflamed tissues. A lack of dedicated tools has hampered their detection and study. We show that a synthesized peptide, MUB40, binds to lactoferrin, the most abundant protein stored in neutrophil-specific and tertiary granules. Lactoferrin is specifically produced by neutrophils among other leukocytes, making MUB40 a specific neutrophil marker. Naive mammalian neutrophils (human, guinea pig, mouse, rabbit) were labeled by fluorescent MUB40 conjugates (-Cy5, Dylight405). A peptidase-resistant retro-inverso MUB40 (RI-MUB40) was synthesized and its lactoferrin-binding property validated. Neutrophil lactoferrin secretion during in vitro Shigella infection was assessed with RI-MUB40-Cy5 using live cell microscopy. Systemically administered RI-MUB40-Cy5 accumulated at sites of inflammation in a mouse arthritis inflammation model in vivo and showed usefulness as a potential tool for inflammation detection using non-invasive imaging. Improving neutrophil detection with the universal and specific MUB40 marker will aid the study of broad ranges of inflammatory diseases.


Assuntos
Carbocianinas/química , Corantes Fluorescentes/química , Inflamação/diagnóstico , Lactoferrina/análise , Neutrófilos/imunologia , Peptídeos/química , Adulto , Animais , Biomarcadores/análise , Disenteria Bacilar/complicações , Disenteria Bacilar/diagnóstico , Disenteria Bacilar/imunologia , Disenteria Bacilar/microbiologia , Feminino , Cobaias , Humanos , Inflamação/complicações , Inflamação/imunologia , Inflamação/microbiologia , Lactoferrina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neutrófilos/microbiologia , Coelhos , Shigella/imunologia
19.
Proc Natl Acad Sci U S A ; 115(2): E283-E291, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29279402

RESUMO

Colonization by Streptococcus gallolyticus subsp. gallolyticus (SGG) is strongly associated with the occurrence of colorectal cancer (CRC). However, the factors leading to its successful colonization are unknown, and whether SGG influences the oncogenic process or benefits from the tumor-prone environment to prevail remains an open question. Here, we elucidate crucial steps that explain how CRC favors SGG colonization. By using mice genetically prone to CRC, we show that SGG colonization is 1,000-fold higher in tumor-bearing mice than in normal mice. This selective advantage occurs at the expense of resident intestinal enterococci. An SGG-specific locus encoding a bacteriocin ("gallocin") is shown to kill enterococci in vitro. Importantly, bile acids strongly enhance this bacteriocin activity in vivo, leading to greater SGG colonization. Constitutive activation of the Wnt pathway, one of the earliest signaling alterations in CRC, and the decreased expression of the bile acid apical transporter gene Slc10A2, as an effect of the Apc founding mutation, may thereby sustain intestinal colonization by SGG. We conclude that CRC-specific conditions promote SGG colonization of the gut by replacing commensal enterococci in their niche.


Assuntos
Neoplasias Colorretais/metabolismo , Trato Gastrointestinal/microbiologia , Streptococcus gallolyticus/fisiologia , Adenoma , Animais , Bacteriocinas/genética , Bacteriocinas/metabolismo , Ácidos e Sais Biliares/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Simportadores/genética , Simportadores/metabolismo
20.
Elife ; 62017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29231810

RESUMO

Disruption of the sumoylation/desumoylation equilibrium is associated with several disease states such as cancer and infections, however the mechanisms regulating the global SUMO balance remain poorly defined. Here, we show that infection by Shigella flexneri, the causative agent of human bacillary dysentery, switches off host sumoylation during epithelial cell infection in vitro and in vivo and that this effect is mainly mediated by a calcium/calpain-induced cleavage of the SUMO E1 enzyme SAE2, thus leading to sumoylation inhibition. Furthermore, we describe a mechanism by which Shigella promotes its own invasion by altering the sumoylation state of RhoGDIα, a master negative regulator of RhoGTPase activity and actin polymerization. Together, our data suggest that SUMO modification is essential to restrain pathogenic bacterial entry by limiting cytoskeletal rearrangement induced by bacterial effectors. Moreover, these findings identify calcium-activated calpains as powerful modulators of cellular sumoylation levels with potentially broad implications in several physiological and pathological situations.


Assuntos
Cálcio/metabolismo , Calpaína/metabolismo , Disenteria Bacilar/microbiologia , Interações Hospedeiro-Patógeno , Shigella flexneri/patogenicidade , Enzimas Ativadoras de Ubiquitina/metabolismo , Disenteria Bacilar/metabolismo , Disenteria Bacilar/patologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Células HeLa , Humanos , Proteólise , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...