Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Gut ; 72(6): 1143-1154, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36585238

RESUMO

OBJECTIVE: Colorectal cancer (CRC) is the third most diagnosed cancer, and requires surgical resection and reconnection, or anastomosis, of the remaining bowel to re-establish intestinal continuity. Anastomotic leak (AL) is a major complication that increases mortality and cancer recurrence. Our objective is to assess the causal role of gut microbiota in anastomotic healing. DESIGN: The causal role of gut microbiota was assessed in a murine AL model receiving faecal microbiota transplantation (FMT) from patients with CRC collected before surgery and who later developed or not, AL. Anastomotic healing and gut barrier integrity were assessed after surgery. Bacterial candidates implicated in anastomotic healing were identified using 16S rRNA gene sequencing and were isolated from faecal samples to be tested both in vitro and in vivo. RESULTS: Mice receiving FMT from patients that developed AL displayed poor anastomotic healing. Profiling of gut microbiota of patients and mice after FMT revealed correlations between healing parameters and the relative abundance of Alistipes onderdonkii and Parabacteroides goldsteinii. Oral supplementation with A. onderdonkii resulted in a higher rate of leaks in mice, while gavage with P. goldsteinii improved healing by exerting an anti-inflammatory effect. Patients with AL and mice receiving FMT from AL patients presented upregulation of mucosal MIP-1α, MIP-2, MCP-1 and IL-17A/F before surgery. Retrospective analysis revealed that patients with AL present higher circulating neutrophil and monocyte counts before surgery. CONCLUSION: Gut microbiota plays an important role in surgical colonic healing in patients with CRC. The impact of these findings may extend to a vast array of invasive gastrointestinal procedures.


Assuntos
Neoplasias Colorretais , Microbioma Gastrointestinal , Camundongos , Animais , Citocinas , Microbioma Gastrointestinal/fisiologia , Estudos Retrospectivos , RNA Ribossômico 16S , Anastomose Cirúrgica/efeitos adversos , Fístula Anastomótica/microbiologia , Neoplasias Colorretais/cirurgia
2.
Cancers (Basel) ; 14(11)2022 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-35681775

RESUMO

Milk fat globule-epidermal growth factor-8 (MFG-E8) is a glycoprotein secreted by different cell types, including apoptotic cells and activated macrophages. MFG-E8 is highly expressed in a variety of cancers and is classically associated with tumor growth and poor patient prognosis through reprogramming of macrophages into the pro-tumoral/pro-angiogenic M2 phenotype. To date, correlations between levels of MFG-E8 and patient survival in prostate and renal cancers remain unclear. Here, we quantified MFG-E8 and CD68/CD206 expression by immunofluorescence staining in tissue microarrays constructed from renal (n = 190) and prostate (n = 274) cancer patient specimens. Percentages of MFG-E8-positive surface area were assessed in each patient core and Kaplan-Meier analyses were performed accordingly. We found that MFG-E8 was expressed more abundantly in malignant regions of prostate tissue and papillary renal cell carcinoma but was also increased in the normal adjacent regions in clear cell renal carcinoma. In addition, M2 tumor-associated macrophage staining was increased in the normal adjacent tissues compared to the malignant areas in renal cancer patients. Overall, high tissue expression of MFG-E8 was associated with less disease progression and better survival in prostate and renal cancer patients. Our observations provide new insights into tumoral MFG-E8 content and macrophage reprogramming in cancer.

3.
J Invest Dermatol ; 137(9): 2005-2013, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28526301

RESUMO

Macrophages are essential for tissue repair. They have a crucial role in cutaneous wound healing, participating actively in the inflammation phase of the process. Unregulated macrophage activation may, however, represent a source of excessive inflammation, leading to abnormal wound healing and hypertrophic scars. Our research group has shown that apoptotic endothelial and epithelial cells secrete MFG-E8, which has the ability to reprogram macrophages from an M1 (proinflammatory) to an M2 (anti-inflammatory, pro-repair) phenotype. Hence, we tested whether modulation of macrophage reprogramming would promote tissue repair. Using a mouse model of wound healing, we showed that the presence and/or addition of MFG-E8 favors wound closure associated with an increase in CD206-positive cells and basic fibroblast growth factor production in healing tissues. More importantly, adoptive transfer of ex vivo MFG-E8-treated macrophages promoted wound closure. We also observed that MFG-E8-treated macrophages produced basic fibroblast growth factor that is responsible for fibroblast migration and proliferation. Taken together, our results strongly suggest that MFG-E8 plays a key role in macrophage reprogramming in tissue healing through induction of an anti-inflammatory M2 phenotype and basic fibroblast growth factor production, leading to fibroblast migration and wound closure.


Assuntos
Antígenos de Superfície/metabolismo , Apoptose/fisiologia , Proteínas do Leite/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Cicatrização/fisiologia , Animais , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Distribuição Aleatória , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/patologia
4.
J Leukoc Biol ; 100(5): 1135-1146, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27260955

RESUMO

Mediators released by apoptotic renal resident cells play a crucial role in modification of the inflammatory microenvironment. We have demonstrated that milk fat globule epidermal growth factor 8 (MFG-E8) is released by apoptotic cells, which results in reduced proinflammatory cytokine production by macrophages. The present study was designed to study the role of MFG-E8 on the modulation of tissue damage and macrophage phenotype in a renal inflammatory model, unilateral ureteral obstruction (UUO). C57BL/6 WT or MFG-E8 KO mice underwent ureteral ligation for 3, 7, and 14 d to evaluate renal injury. MFG-E8 (30 µg/kg) or vehicle was also administered i.p. MFG-E8 administration reduced kidney damage and fibrosis compared with control, whereas its absence in MFG-E8 KO mice was associated with more severe disease. Moreover, MFG-E8 administration was associated with decreased inflammasome activation in the kidney. Furthermore, adoptive transfer of MFG-E8-stimulated macrophages reduced activation of inflammasome and tissue damage. In all cases, both the systemic administration of MFG-E8 and MFG-E8-treated macrophages promoted accumulation of anti-inflammatory CD206+ macrophages. We propose that the protective role of MFG-E8 is mediated through anti-inflammatory macrophage reprogramming which results in decreased inflammasome activation, preventing severe tissue damage. These data provide valuable insight for identification of MFG-E8 as a novel target in modulation of inflammatory diseases.


Assuntos
Antígenos de Superfície/imunologia , Inflamassomos/imunologia , Macrófagos/imunologia , Proteínas do Leite/imunologia , Nefrite/imunologia , Animais , Antígenos de Superfície/genética , Antígenos de Superfície/uso terapêutico , Apoptose , Microambiente Celular , Colágeno/análise , Imunoterapia Adotiva , Inflamassomos/efeitos dos fármacos , Rim/química , Rim/imunologia , Rim/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/transplante , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Leite/genética , Proteínas do Leite/uso terapêutico , Nefrite/etiologia , Nefrite/patologia , Nefrite/terapia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Obstrução Ureteral/complicações
5.
J Biol Chem ; 286(49): 42494-42503, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-21965665

RESUMO

ß(2)-Glycoprotein I (ß(2)GPI) is an abundant plasma protein that binds to the surface of cells and particles expressing negatively charged lipids, but its physiological role remains unknown. Antibodies to ß(2)GPI are found in patients with anti-phospholipid syndrome, a systemic autoimmune disease associated with vascular thrombosis and pregnancy morbidity. Although it has been suggested that anti-ß(2)GPI antibodies activate endothelial cells and monocytes by signaling through TLR4, it is unclear how anti-ß(2)GPI antibodies and/or ß(2)GPI interact with TLR4. A number of mammalian proteins (termed "endogenous Toll-like receptor (TLR) ligands") have been reported to bind to TLR4, but, in most cases, subsequent studies have shown that LPS interaction with these proteins is responsible for TLR activation. We hypothesized that, like other endogenous TLR ligands, ß(2)GPI interacts specifically with LPS and that this interaction is responsible for apparent TLR4 activation by ß(2)GPI. Here, we show that both LPS and TLR4 are required for ß(2)GPI to bind to and activate macrophages. Untreated ß(2)GPI stimulated TNF-α production in TLR4-sufficient (but not TLR4-deficient) macrophages. In contrast, neither polymyxin B-treated nor delipidated ß(2)GPI stimulated TNF-α production. Furthermore, ß(2)GPI bound to LPS in a specific and dose-dependent manner. Finally, untreated ß(2)GPI bound to the surface of TLR4-sufficient (but not TLR4-deficient) macrophages. Polymyxin B treatment of ß(2)GPI abolished macrophage binding. Our findings suggest a potential new biological activity for ß(2)GPI as a protein that interacts specifically with LPS and point to the need to evaluate newly discovered endogenous TLR ligands for potential interactions with LPS.


Assuntos
Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Receptor 4 Toll-Like/metabolismo , beta 2-Glicoproteína I/metabolismo , Animais , Endotoxinas/metabolismo , Feminino , Regulação da Expressão Gênica , Imunidade Inata , Ligantes , Lipídeos/química , Lipopolissacarídeos/química , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/métodos , Polimixina B/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
6.
J Biol Chem ; 283(40): 27220-9, 2008 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-18658137

RESUMO

Proteolysis of extracellular matrix components and the production of cryptic bioactive factors play key roles in vascular remodeling. We showed previously that extracellular matrix proteolysis is triggered by the apoptosis of endothelial cells (EC), resulting in the release of an anti-apoptotic C-terminal fragment of endorepellin (LG3). Here, we characterize the endorepellin-cleaving proteases released by apoptotic EC using a multifaceted proteomics strategy. Cathepsin L (CathL), a cysteine protease known to be associated with cardiovascular disease progression in animal models and humans, was isolated from medium conditioned by apoptotic EC. CathL cleaved recombinant endorepellin in vitro, leading to LG3 release. Inhibition of CathL activity in EC exposed to pro-apoptotic stimuli prevented LG3 release without modulating the development of apoptosis in EC. Inhibition of caspase-3 activation in EC with the biochemical inhibitor DEVD-fluoromethyl ketone or small interfering RNAs concomitantly prevented CathL release by EC, LG3 production, and the development of paracrine anti-apoptotic activity. These data demonstrate that caspase-3 activation is a novel pathway of importance for triggering extracellular CathL release and the cleavage of extracellular matrix components.


Assuntos
Apoptose/fisiologia , Caspase 3/metabolismo , Catepsinas/metabolismo , Cisteína Endopeptidases/metabolismo , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Doenças Cardiovasculares/metabolismo , Catepsina L , Linhagem Celular , Modelos Animais de Doenças , Humanos , Comunicação Parácrina/fisiologia , Peptídeos/metabolismo , RNA Interferente Pequeno
7.
FASEB J ; 18(6): 705-7, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14977881

RESUMO

Increased endothelial apoptosis and decreased apoptosis of vascular smooth muscle cells (VSMC) are central to initiation of myo-intimal thickening. We hypothesized that apoptosis of endothelial cells (EC) induces the release of anti-apoptotic mediator(s) active on VSMC. We found that serum-free medium conditioned by apoptotic EC decreases apoptosis of VSMC compared with fresh serum-free medium. Inhibition of endothelial apoptosis during conditioning with a pan-caspase inhibitor ZVAD-FMK blocked the release of the anti-apoptotic factor(s) active on VSMC. VSMC exposed to serum-free medium conditioned by apoptotic EC showed increased ERK 1/2 phosphorylation, enhanced Bcl-xl expression, and inhibition of p53 expression. Fractionation of the conditioned medium followed by mass spectral analysis identified one bioactive component as a C-terminal fragment of the domain V of perlecan. Serum-free medium supplemented with either a synthetic peptide containing the EGF motif of the domain V of perlecan or chondroitin 4-sulfate, a glycosaminoglycan anchored on the domain V of perlecan, increased ERK 1/2 phosphorylation and Bcl-xl protein levels while inhibiting apoptosis of VSMC. These results suggest that a proteolytic activity developing downstream of activated caspases in apoptotic EC initiates degradation of pericellular proteoglycans and liberation of bioactive fragments with a robust impact on inhibition of VSMC apoptosis.


Assuntos
Apoptose , Caspases/metabolismo , Endotélio Vascular/enzimologia , Músculo Liso Vascular/metabolismo , Comunicação Parácrina , Animais , Fatores Biológicos/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Proteoglicanas de Heparan Sulfato/química , Proteoglicanas de Heparan Sulfato/fisiologia , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Músculo Liso Vascular/citologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Proteína Supressora de Tumor p53/metabolismo , Proteína bcl-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA