Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cell Mol Gastroenterol Hepatol ; 18(1): 53-70, 2024 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-38438014

RESUMO

BACKGROUND & AIMS: The abdominal discomfort experienced by patients with colitis may be attributable in part to the presence of small intestinal dysmotility, yet mechanisms linking colonic inflammation with small-bowel motility remain largely unexplored. We hypothesize that colitis results in small intestinal hypomotility owing to a loss of enteroendocrine cells (EECs) within the small intestine that can be rescued using serotonergic-modulating agents. METHODS: Male C57BL/6J mice, as well as mice that overexpress (EECOVER) or lack (EECDEL) NeuroD1+ enteroendocrine cells, were exposed to dextran sulfate sodium (DSS) colitis (2.5% or 5% for 7 days) and small intestinal motility was assessed by 70-kilodalton fluorescein isothiocyanate-dextran fluorescence transit. EEC number and differentiation were evaluated by immunohistochemistry, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling staining, and quantitative reverse-transcriptase polymerase chain reaction. Mice were treated with the 5-hydroxytryptamine receptor 4 agonist prucalopride (5 mg/kg orally, daily) to restore serotonin signaling. RESULTS: DSS-induced colitis was associated with a significant small-bowel hypomotility that developed in the absence of significant inflammation in the small intestine and was associated with a significant reduction in EEC density. EEC loss occurred in conjunction with alterations in the expression of key serotonin synthesis and transporter genes, including Tph1, Ddc, and Slc6a4. Importantly, mice overexpressing EECs revealed improved small intestinal motility, whereas mice lacking EECs had worse intestinal motility when exposed to DSS. Finally, treatment of DSS-exposed mice with the 5-hydroxytryptamine receptor 4 agonist prucalopride restored small intestinal motility and attenuated colitis. CONCLUSIONS: Experimental DSS colitis induces significant small-bowel dysmotility in mice owing to enteroendocrine loss that can be reversed by genetic modulation of EEC or administering serotonin analogs, suggesting novel therapeutic approaches for patients with symptomatic colitis.

2.
Am J Physiol Gastrointest Liver Physiol ; 325(1): G23-G41, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37120853

RESUMO

Necrotizing enterocolitis (NEC) is the leading cause of morbidity and mortality in premature infants. One of the most devastating complications of NEC is the development of NEC-induced brain injury, which manifests as impaired cognition that persists beyond infancy and which represents a proinflammatory activation of the gut-brain axis. Given that oral administration of the human milk oligosaccharides (HMOs) 2'-fucosyllactose (2'-FL) and 6'-sialyslactose (6'-SL) significantly reduced intestinal inflammation in mice, we hypothesized that oral administration of these HMOs would reduce NEC-induced brain injury and sought to determine the mechanisms involved. We now show that the administration of either 2'-FL or 6'-SL significantly attenuated NEC-induced brain injury, reversed myelin loss in the corpus callosum and midbrain of newborn mice, and prevented the impaired cognition observed in mice with NEC-induced brain injury. In seeking to define the mechanisms involved, 2'-FL or 6'-SL administration resulted in a restoration of the blood-brain barrier in newborn mice and also had a direct anti-inflammatory effect on the brain as revealed through the study of brain organoids. Metabolites of 2'-FL were detected in the infant mouse brain by nuclear magnetic resonance (NMR), whereas intact 2'-FL was not. Strikingly, the beneficial effects of 2'-FL or 6'-SL against NEC-induced brain injury required the release of the neurotrophic factor brain-derived neurotrophic factor (BDNF), as mice lacking BDNF were not protected by these HMOs from the development of NEC-induced brain injury. Taken in aggregate, these findings reveal that the HMOs 2'-FL and 6'-SL interrupt the gut-brain inflammatory axis and reduce the risk of NEC-induced brain injury.NEW & NOTEWORTHY This study reveals that the administration of human milk oligosaccharides, which are present in human breast milk, can interfere with the proinflammatory gut-brain axis and prevent neuroinflammation in the setting of necrotizing enterocolitis, a major intestinal disorder seen in premature infants.


Assuntos
Lesões Encefálicas , Disfunção Cognitiva , Enterocolite Necrosante , Humanos , Recém-Nascido , Lactente , Feminino , Animais , Camundongos , Leite Humano/metabolismo , Fator Neurotrófico Derivado do Encéfalo , Doenças Neuroinflamatórias , Enterocolite Necrosante/etiologia , Oligossacarídeos/farmacologia , Oligossacarídeos/uso terapêutico , Oligossacarídeos/análise , Disfunção Cognitiva/prevenção & controle , Disfunção Cognitiva/complicações , Lesões Encefálicas/complicações , Lesões Encefálicas/metabolismo
3.
Am J Physiol Gastrointest Liver Physiol ; 323(3): G265-G282, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35819175

RESUMO

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in premature infants and is steadily rising in frequency. Patients who develop NEC have a very high mortality, illustrating the importance of developing novel prevention or treatment approaches. We and others have shown that NEC arises in part from exaggerated signaling via the bacterial receptor, Toll-like receptor 4 (TLR4) on the intestinal epithelium, leading to widespread intestinal inflammation and intestinal ischemia. Strategies that limit the extent of TLR4 signaling, including the administration of amniotic fluid, can reduce NEC development in mouse and piglet models. We now seek to test the hypothesis that a secretome derived from amnion-derived cells can prevent or treat NEC in preclinical models of this disease via a process involving TLR4 inhibition. In support of this hypothesis, we show that the administration of this secretome, named ST266, to mice or piglets can prevent and treat experimental NEC. The protective effects of ST266 occurred in the presence of marked TLR4 inhibition in the intestinal epithelium of cultured epithelial cells, intestinal organoids, and human intestinal samples ex vivo, independent of epidermal growth factor. Strikingly, RNA-seq analysis of the intestinal epithelium in mice reveals that the ST266 upregulates critical genes associated with gut remodeling, intestinal immunity, gut differentiation. and energy metabolism. These findings show that the amnion-derived secretome ST266 can prevent and treat NEC, suggesting the possibility of novel therapeutic approaches for patients with this devastating disease.NEW & NOTEWORTHY This work provides hope for children who develop NEC, a devastating disease of premature infants that is often fatal, by revealing that the secreted product of amniotic progenitor cells (called ST266) can prevent or treat NEC in mice, piglet, and "NEC-in-a-dish" models of this disease. Mechanistically, ST266 prevented bacterial signaling, and a detailed transcriptomic analysis revealed effects on gut differentiation, immunity, and metabolism. Thus, an amniotic secretome may offer novel approaches for NEC.


Assuntos
Enterocolite Necrosante , Células-Tronco Multipotentes , Secretoma , Âmnio/citologia , Animais , Modelos Animais de Doenças , Enterocolite Necrosante/prevenção & controle , Mucosa Intestinal/metabolismo , Camundongos , Células-Tronco Multipotentes/metabolismo , Suínos , Receptor 4 Toll-Like/metabolismo
4.
Br J Nutr ; 128(6): 1050-1063, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34632971

RESUMO

Necrotising enterocolitis (NEC) is a devastating gastrointestinal disease of prematurity that typically develops after the administration of infant formula, suggesting a link between nutritional components and disease development. One of the most significant complications that develops in patients with NEC is severe lung injury. We have previously shown that the administration of a nutritional formula that is enriched in pre-digested Triacylglyceride that do not require lipase action can significantly reduce the severity of NEC in a mouse model. We now hypothesise that this 'pre-digested fat (PDF) system' may reduce NEC-associated lung injury. In support of this hypothesis, we now show that rearing newborn mice on a nutritional formula based on the 'PDF system' promotes lung development, as evidenced by increased tight junctions and surfactant protein expression. Mice that were administered this 'PDF system' were significantly less vulnerable to the development of NEC-induced lung inflammation, and the administration of the 'PDF system' conferred lung protection. In seeking to define the mechanisms involved, the administration of the 'PDF system' significantly enhanced lung maturation and reduced the production of reactive oxygen species (ROS). These findings suggest that the PDF system protects the development of NEC-induced lung injury through effects on lung maturation and reduced ROS in the lung and also increases lung maturation in non-NEC mice.


Assuntos
Enterocolite Necrosante , Lesão Pulmonar , Animais , Camundongos , Enterocolite Necrosante/etiologia , Animais Recém-Nascidos , Espécies Reativas de Oxigênio , Lesão Pulmonar/complicações , Lesão Pulmonar/metabolismo , Alimentos Formulados , Modelos Animais de Doenças
5.
Sci Transl Med ; 13(612): eabg3459, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34550727

RESUMO

Necrotizing enterocolitis (NEC) is a devastating disease of premature infants, whose pathogenesis remains incompletely understood, although activation of the Gram-negative bacterial receptor Toll-like receptor 4 (TLR4) on the intestinal epithelium plays a critical role. Patients with NEC typically display gastrointestinal dysmotility before systemic disease is manifest, suggesting that dysmotility could drive NEC development. Both intestinal motility and inflammation are governed by the enteric nervous system, a network of enteric neurons and glia. We hypothesized here that enteric glia loss in the premature intestine could lead to dysmotility, exaggerated TLR4 signaling, and NEC development. We found that intestinal motility is reduced early in NEC in mice, preceding the onset of intestinal inflammation, whereas pharmacologic restoration of intestinal motility reduced NEC severity. Ileal samples from mouse, piglet, and human NEC revealed enteric glia depletion, and glia-deficient mice (Plp1ΔDTR, Sox10ΔDTR, and BdnfΔDTR) showed increased NEC severity compared with wild-type mice. Mice lacking TLR4 on enteric glia (Sox10-Tlr4ko) did not show NEC-induced enteric glia depletion and were protected from NEC. Mechanistically, brain-derived neurotrophic factor (BDNF) from enteric glia restrained TLR4 signaling on the intestine to prevent NEC. BDNF was reduced in mouse and human NEC, and BDNF administration reduced both TLR4 signaling and NEC severity in enteric glia­deficient mice. Last, we identified an agent (J11) that enhanced enteric glial BDNF release, inhibited intestinal TLR4, restored motility, and prevented NEC in mice. Thus, enteric glia loss might contribute to NEC through intestinal dysmotility and increased TLR4 activation, suggesting enteric glia therapies for this disorder.


Assuntos
Enterocolite Necrosante , Receptor 4 Toll-Like , Humanos , Recém-Nascido , Neuroglia , Receptor 4 Toll-Like/genética , Animais , Camundongos
6.
Nat Commun ; 12(1): 1042, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33589625

RESUMO

Necrotizing enterocolitis (NEC) is a disease of premature infants characterized by acute intestinal necrosis. Current dogma suggests that NEC develops in response to post-natal dietary and bacterial factors, and so a potential role for in utero factors in NEC remains unexplored. We now show that during pregnancy, administration of a diet rich in the aryl hydrocarbon receptor (AHR) ligand indole-3-carbinole (I3C), or of breast milk, activates AHR and prevents NEC in newborn mice by reducing Toll-like receptor 4 (TLR4) signaling in the newborn gut. Protection from NEC requires activation of AHR in the intestinal epithelium which is reduced in mouse and human NEC, and is independent of leukocyte activation. Finally, we identify an AHR ligand ("A18") that limits TLR4 signaling in mouse and human intestine, and prevents NEC in mice when administered during pregnancy. In summary, AHR signaling is critical in NEC development, and maternally-delivered, AHR-based therapies may alleviate NEC.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Enterocolite Necrosante/genética , Indóis/administração & dosagem , Leite Humano/fisiologia , Receptores de Hidrocarboneto Arílico/genética , Receptor 4 Toll-Like/genética , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/agonistas , Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/imunologia , Dieta/métodos , Modelos Animais de Doenças , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/patologia , Enterocolite Necrosante/prevenção & controle , Feminino , Regulação da Expressão Gênica , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Ligantes , Exposição Materna , Camundongos , Gravidez , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/imunologia , Transdução de Sinais , Suínos , Receptor 4 Toll-Like/imunologia
7.
Sci Transl Med ; 13(575)2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33408187

RESUMO

Necrotizing enterocolitis (NEC) causes acute intestinal necrosis in premature infants and is associated with severe neurological impairment. In NEC, Toll-like receptor 4 is activated in the intestinal epithelium, and NEC-associated brain injury is characterized by microglial activation and white matter loss through mechanisms that remain unclear. We now show that the brains of mice and humans with NEC contained CD4+ T lymphocytes that were required for the development of brain injury. Inhibition of T lymphocyte influx into the brains of neonatal mice with NEC reduced inflammation and prevented myelin loss. Adoptive intracerebroventricular delivery of gut T lymphocytes from mice with NEC into Rag1 -/- recipient mice lacking CD4+ T cells resulted in brain injury. Brain organoids derived from mice with or without NEC and from human neuronal progenitor cells revealed that IFN-γ release by CD4+ T lymphocytes induced microglial activation and myelin loss in the organoids. IFN-γ knockdown in CD4+ T cells derived from mice with NEC abrogated the induction of NEC-associated brain injury after adoptive transfer to naïve Rag1 -/- recipient mice. T cell receptor sequencing revealed that NEC mouse brain-derived T lymphocytes shared homology with gut T lymphocytes from NEC mice. Intraperitoneal injection of NEC gut-derived CD4+ T lymphocytes into naïve Rag1 -/- recipient mice induced brain injury, suggesting that gut-derived T lymphocytes could mediate neuroinflammation in NEC. These findings indicate that NEC-associated brain injury may be induced by gut-derived IFN-γ-releasing CD4+ T cells, suggesting that early management of intestinal inflammation in children with NEC could improve neurological outcomes.


Assuntos
Enterocolite Necrosante , Animais , Encéfalo , Inflamação , Mucosa Intestinal , Camundongos , Linfócitos T Reguladores
9.
Pediatr Res ; 89(1): 91-101, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32221473

RESUMO

BACKGROUND: Necrotizing enterocolitis (NEC) develops through exaggerated toll-like receptor 4 (TLR4) signaling in the intestinal epithelium. Breast milk is rich in non-digestible oligosaccharides and prevents NEC through unclear mechanisms. We now hypothesize that the human milk oligosaccharides 2'-fucosyllactose (2'-FL) and 6'-sialyllactose (6'-SL) can reduce NEC through inhibition of TLR4 signaling. METHODS: NEC was induced in newborn mice and premature piglets and infant formula was supplemented with 2'-FL, 6'-SL, or lactose. Intestinal tissue was obtained at surgical resection. HMO inhibition of TLR4 was assessed in IEC-6 enterocytes, mice, and human tissue explants and via in silico modeling. RESULTS: Supplementation of infant formula with either 2'-FL and/or 6'-SL, but not the parent sugar lactose, reduced NEC in mice and piglets via reduced apoptosis, inflammation, weight loss, and histological appearance. Mechanistically, both 2'-FL and 6'-SL, but not lactose, reduced TLR4-mediated nuclear factor kappa light-chain enhancer of activated B cells (NF-kB) inflammatory signaling in the mouse and human intestine. Strikingly, in silico modeling revealed 2'-FL and 6'-SL, but not lactose, to dock into the binding pocket of the TLR4-MD2 complex, explaining their ability to inhibit TLR4 signaling. CONCLUSIONS: 2'-FL and 6'-SL, but not lactose, prevent NEC in mice and piglet models and attenuate NEC inflammation in the human ileum, in part through TLR4 inhibition. IMPACT: Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants that occurs in the setting of bacterial colonization of the gut and administration of formula feeds and activation by the innate immune receptor toll-like receptor 4 (TLR4). Breast milk prevents NEC through unclear mechanisms. We now show that breast milk-enriched human milk oligosaccharides (HMOs) that are derived from lactose prevent NEC through inhibition of TLR4. The human milk oligosaccharides 2'-FL and 6'-SL, but not the backbone sugar lactose, prevent NEC in mice and piglets. 2'-FL and 6'-SL but not lactose inhibited TLR4 signaling in cultured enterocytes, in enteroids derived from mouse intestine, and in human intestinal explants obtained at the time of surgical resection for patients with NEC. In seeking the mechanisms involved, 2'-FL and 6'-SL but not lactose were found to directly bind to TLR4, explaining the inhibition and protection against NEC. These findings may impact clinical practice by suggesting that administration of HMOs could serve as a preventive strategy for premature infants at risk for NEC development.


Assuntos
Enterocolite Necrosante/prevenção & controle , Íleo/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Lactose/análogos & derivados , Leite Humano/química , Receptor 4 Toll-Like/antagonistas & inibidores , Trissacarídeos/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/patologia , Humanos , Íleo/imunologia , Íleo/metabolismo , Íleo/patologia , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Lactose/isolamento & purificação , Lactose/farmacologia , Camundongos , Simulação de Acoplamento Molecular , Transdução de Sinais , Sus scrofa , Receptor 4 Toll-Like/metabolismo , Trissacarídeos/isolamento & purificação , Redução de Peso/efeitos dos fármacos
10.
Cell Mol Gastroenterol Hepatol ; 9(3): 403-423, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31756560

RESUMO

BACKGROUND & AIMS: Necrotizing enterocolitis (NEC) is a devastating disease of premature infants characterized by Toll-like receptor 4 (TLR4)-dependent intestinal inflammation and enterocyte death. Given that necroptosis is a proinflammatory cell death process that is linked to bacterial signaling, we investigated its potential role in NEC, and the mechanisms involved. METHODS: Human and mouse NEC intestine were analyzed for necroptosis gene expression (ie, RIPK1, RIPK3, and MLKL), and protein activation (phosphorylated RIPK3). To evaluate a potential role for necroptosis in NEC, the effects of genetic (ie, Ripk3 knockout or Mlkl knockout) or pharmacologic (ie, Nec1s) inhibition of intestinal inflammation were assessed in a mouse NEC model, and a possible upstream role of TLR4 was assessed in Tlr4-deficient mice. The NEC-protective effects of human breast milk and its constituent milk oligosaccharides on necroptosis were assessed in a NEC-in-a-dish model, in which mouse intestinal organoids were cultured as either undifferentiated or differentiated epithelium in the presence of NEC bacteria and hypoxia. RESULTS: Necroptosis was activated in the intestines of human and mouse NEC in a TLR4-dependent manner, and was up-regulated specifically in differentiated epithelium of the immature ileum. Inhibition of necroptosis genetically and pharmacologically reduced intestinal-epithelial cell death and mucosal inflammation in experimental NEC, and ex vivo in the NEC-in-a-dish system. Strikingly, the addition of human breast milk, or the human milk oligosaccharide 2 fucosyllactose in the ex vivo system, reduced necroptosis and inflammation. CONCLUSIONS: Necroptosis is activated in the intestinal epithelium upon TLR4 signaling and is required for NEC development, and explains in part the protective effects of breast milk.


Assuntos
Enterocolite Necrosante/patologia , Enterócitos/patologia , Mucosa Intestinal/patologia , Leite Humano/química , Necroptose/imunologia , Animais , Modelos Animais de Doenças , Enterocolite Necrosante/tratamento farmacológico , Enterocolite Necrosante/genética , Enterocolite Necrosante/imunologia , Enterócitos/efeitos dos fármacos , Enterócitos/imunologia , Feminino , Humanos , Recém-Nascido , Mucosa Intestinal/efeitos dos fármacos , Camundongos , Camundongos Knockout , Necroptose/efeitos dos fármacos , Proteínas Quinases/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Trissacarídeos/farmacologia , Trissacarídeos/uso terapêutico , Regulação para Cima
11.
J Immunol ; 203(11): 3000-3012, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31645418

RESUMO

Angiotensin-converting enzyme 2 (ACE2) is a potent negative regulator capable of restraining overactivation of the renin-angiotensin system, which contributes to exuberant inflammation after bacterial infection. However, the mechanism through which ACE2 modulates this inflammatory response is not well understood. Accumulating evidence indicates that infectious insults perturb ACE2 activity, allowing for uncontrolled inflammation. In the current study, we demonstrate that pulmonary ACE2 levels are dynamically varied during bacterial lung infection, and the fluctuation is critical in determining the severity of bacterial pneumonia. Specifically, we found that a pre-existing and persistent deficiency of active ACE2 led to excessive neutrophil accumulation in mouse lungs subjected to bacterial infection, resulting in a hyperinflammatory response and lung damage. In contrast, pre-existing and persistent increased ACE2 activity reduces neutrophil infiltration and compromises host defense, leading to overwhelming bacterial infection. Further, we found that the interruption of pulmonary ACE2 restitution in the model of bacterial lung infection delays the recovery process from neutrophilic lung inflammation. We observed the beneficial effects of recombinant ACE2 when administered to bacterially infected mouse lungs following an initial inflammatory response. In seeking to elucidate the mechanisms involved, we discovered that ACE2 inhibits neutrophil infiltration and lung inflammation by limiting IL-17 signaling by reducing the activity of the STAT3 pathway. The results suggest that the alteration of active ACE2 is not only a consequence of bacterial lung infection but also a critical component of host defense through modulation of the innate immune response to bacterial lung infection by regulating neutrophil influx.


Assuntos
Inflamação/imunologia , Neutrófilos/imunologia , Peptidil Dipeptidase A/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Enzima de Conversão de Angiotensina 2 , Animais , Modelos Animais de Doenças , Feminino , Imidazóis/administração & dosagem , Imidazóis/farmacologia , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Inflamação/tratamento farmacológico , Inflamação/patologia , Leucina/administração & dosagem , Leucina/análogos & derivados , Leucina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Testes de Sensibilidade Microbiana , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Peptidil Dipeptidase A/deficiência , Peptidil Dipeptidase A/genética , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/efeitos dos fármacos
12.
Shock ; 52(2): 215-223, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30148762

RESUMO

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in premature infants, and is associated with the development of severe lung inflammation. The pathogenesis of NEC-induced lung injury remains unknown, yet infiltrating immune cells may play a role. In support of this possibility, we now show that NEC in mice and humans was associated with the development of profound lung injury that was characterized by an influx of Th17 cells and a reduction in T regulatory lymphocytes (Tregs). Importantly, the adoptive transfer of CD4 T cells isolated from lungs of mice with NEC into the lungs of immune incompetent mice (Rag1 mice) induced profound inflammation in the lung, while the depletion of Tregs exacerbated NEC induced lung injury, demonstrating that imbalance of Th17/Treg in the lung is required for the induction of injury. In seeking to define the mechanisms involved, the selective deletion of toll-like receptor 4 (TLR4) from the Sftpc1 pulmonary epithelial cells reversed lung injury, while TLR4 activation induced the Th17 recruiting chemokine (C-C motif) ligand 25 (CCL25) in the lungs of mice with NEC. Strikingly, the aerosolized inhibition of both CCL25 and TLR4 and the administration of all trans retinoic acid restored Tregs attenuated NEC-induced lung injury. In summary, we show that TLR4 activation in Surfactant protein C-1 (Sftpc1) cells disrupts the Treg/Th17 balance in the lung via CCL25 leading to lung injury after NEC and reveal that inhibition of TLR4 and stabilization of Th17/Treg balance in the neonatal lung may prevent this devastating complication of NEC.


Assuntos
Lesão Pulmonar/metabolismo , Linfócitos/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Quimiocinas CC/metabolismo , Enterocolite/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/metabolismo
13.
Sci Transl Med ; 10(471)2018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30541786

RESUMO

Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease of the premature infant. One of the most important long-term complications observed in children who survive NEC early in life is the development of profound neurological impairments. However, the pathways leading to NEC-associated neurological impairments remain unknown, thus limiting the development of prevention strategies. We have recently shown that NEC development is dependent on the expression of the lipopolysaccharide receptor Toll-like receptor 4 (TLR4) on the intestinal epithelium, whose activation by bacteria in the newborn gut leads to mucosal inflammation. Here, we hypothesized that damage-induced production of TLR4 endogenous ligands in the intestine might lead to activation of microglial cells in the brain and promote cognitive impairments. We identified a gut-brain signaling axis in an NEC mouse model in which activation of intestinal TLR4 signaling led to release of high-mobility group box 1 in the intestine that, in turn, promoted microglial activation in the brain and neurological dysfunction. We further demonstrated that an orally administered dendrimer-based nanotherapeutic approach to targeting activated microglia could prevent NEC-associated neurological dysfunction in neonatal mice. These findings shed light on the molecular pathways leading to the development of NEC-associated brain injury, provide a rationale for early removal of diseased intestine in NEC, and indicate the potential of targeted therapies that protect the developing brain in the treatment of NEC in early childhood.


Assuntos
Encéfalo/patologia , Disfunção Cognitiva/etiologia , Enterocolite Necrosante/complicações , Microglia/patologia , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Administração Oral , Animais , Animais Recém-Nascidos , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Encéfalo/ultraestrutura , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/patologia , Dendrímeros/química , Proteína HMGB1/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Camundongos Endogâmicos C57BL , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Receptor 4 Toll-Like/metabolismo
14.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L17-L31, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28935640

RESUMO

Angiotensin-converting enzyme 2 (ACE2) is a terminal carboxypeptidase with important functions in the renin-angiotensin system and plays a critical role in inflammatory lung diseases. ACE2 cleaves single-terminal residues from several bioactive peptides such as angiotensin II. However, few of its substrates in the respiratory tract have been identified, and the mechanism underlying the role of ACE2 in inflammatory lung disease has not been fully characterized. In an effort to identify biological targets of ACE2 in the lung, we tested its effects on des-Arg9 bradykinin (DABK) in airway epithelial cells on the basis of the hypothesis that DABK is a biological substrate of ACE2 in the lung and ACE2 plays an important role in the pathogenesis of acute lung inflammation partly through modulating DABK/bradykinin receptor B1 (BKB1R) axis signaling. We found that loss of ACE2 function in mouse lung in the setting of endotoxin inhalation led to activation of the DABK/BKB1R axis, release of proinflammatory chemokines such as C-X-C motif chemokine 5 (CXCL5), macrophage inflammatory protein-2 (MIP2), C-X-C motif chemokine 1 (KC), and TNF-α from airway epithelia, increased neutrophil infiltration, and exaggerated lung inflammation and injury. These results indicate that a reduction in pulmonary ACE2 activity contributes to the pathogenesis of lung inflammation, in part because of an impaired ability to inhibit DABK/BKB1R axis-mediated signaling, resulting in more prompt onset of neutrophil infiltration and more severe inflammation in the lung. Our study identifies a biological substrate of ACE2 within the airways, as well as a potential new therapeutic target for inflammatory diseases.


Assuntos
Bradicinina/análogos & derivados , Lipopolissacarídeos/toxicidade , Infiltração de Neutrófilos/imunologia , Peptidil Dipeptidase A/fisiologia , Pneumonia/imunologia , Receptor B1 da Bradicinina/metabolismo , Traqueia/imunologia , Enzima de Conversão de Angiotensina 2 , Animais , Anti-Inflamatórios , Bradicinina/farmacologia , Células Cultivadas , Quimiocina CXCL5/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/efeitos dos fármacos , Pneumonia/induzido quimicamente , Pneumonia/tratamento farmacológico , Pneumonia/metabolismo , Traqueia/efeitos dos fármacos , Traqueia/patologia
15.
J Cell Biochem ; 114(4): 773-81, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23060091

RESUMO

ABBA was reported to be an actin dynamics regulator. However, the molecular mechanism of action of ABBA is still totally obscure. Here, we show that ABBA is ubiquitously expressed in all the examined cultured cells. We found that expression of ABBA in NIH3T3 cells promotes cell spreading. ABBA binds to and markedly promotes cell spreading-induced Rac1 activation. Cell spreading stimulates ABBA activation probably by inducing it tyrosine phosphorylation, which endows ABBA much higher activity to activate Rac1, and attenuates the interaction between ABBA and Rac1. Loss of function suggests that deletion of ABBA in C6-R cells markedly inhibits Rac1 activation and cell spreading; this suggests that and the interaction between ABBA and activated Rac1 is required for ABBA-promoted cell spreading. Taken together, our results indicate that ABBA is activated in response to cell spreading, which markedly promotes cell spreading, and ABBA is required for Rac1 activation and cell spreading.


Assuntos
Movimento Celular , Fibronectinas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Neuropeptídeos/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Forma Celular , Ativação Enzimática , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Proteínas de Membrana/genética , Camundongos , Proteínas dos Microfilamentos/genética , Células NIH 3T3 , Proteínas de Neoplasias/genética , Neuropeptídeos/genética , Fosforilação , Ligação Proteica , Mapeamento de Interação de Proteínas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transfecção , Tirosina/metabolismo , Proteínas rac de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP
16.
Peptides ; 33(1): 44-51, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22115565

RESUMO

BmKbpp is a novel cationic and α-helical peptide from the Chinese scorpion Mesobuthus martensii Karsch, of which function or biological activity has not been characterized so far. Here we showed that BmKbpp possesses strong antimicrobial activity against both Gram-positive and Gram-negative bacteria with a MIC range from 2.3 µM to 68.2 µM for the majority of tested bacteria. BmKbpp also inhibits the growth of tested fungi with an IC50 range from 0.2 µM to 3.1 µM. Because BmKbpp potently inhibits the growth of some antibiotics-resistant pathogens, and shows very weak hemolytic activity, it has considerable potentials for therapeutic applications. Moreover, we found that BmKbpp markedly inhibits the superoxide production in granulocytes or HL-60 cells at the concentrations of submicromolar level; this suggests that BmKbpp can act as a signaling molecule involving innate immune regulation at low concentrations. The C-terminal region of BmKbpp (BmKbpp-C) shows 72% similarity to the peptide K-12, a bradykinin-potentiating peptide. We found that both BmKbpp and BmKbpp-C possess bradykinin-potentiating activity, and the activity of BmKbpp-C is stronger than that of BmKbpp. PCR amplification for the genomic gene of BmBpp showed that it is not a continuous sequence in the genome; it suggests that BmKbpp could come from a recombination event in transcript level. Taken together, our data suggest that multi-functionalization of a single peptide, which is probably mediated by trans-splicing, could be a new mechanism for the functional diversification of scorpion venom peptides.


Assuntos
Venenos de Escorpião/genética , Venenos de Escorpião/farmacologia , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Bradicinina/metabolismo , Relação Dose-Resposta a Droga , Bactérias Aeróbias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Células HL-60/efeitos dos fármacos , Células HL-60/metabolismo , Hemolíticos/farmacologia , Humanos , Concentração Inibidora 50 , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , NADPH Oxidases/metabolismo , Splicing de RNA , Venenos de Escorpião/química , Escorpiões/química , Homologia de Sequência de Aminoácidos , Superóxidos/metabolismo
17.
Peptides ; 25(2): 143-50, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15062994

RESUMO

The scorpion venom peptides with no disulfide bridge are rarely identified and poorly characterized so far. Here, we report the identification and characterization of four novel disulfide-bridge-free venom peptides (BmKa1, BmKa2, BmKb1 and BmKn2) from Buthus martensii Kasch. BmKa1 and BmKa2 are very acidic and hydrophilic, showing no any similarity to other proteins, whereas BmKb1 and BmKn2 both are basic, alpha-helical peptide with an amidated C-terminus, showing a little homology with other peptides. Functional tests with synthetic peptide showed that BmKn2 has strong antimicrobial activity against both Gram-positive and Gram-negative bacteria, whereas BmKb1 has weak activity in inhibiting the growth of these bacteria.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Biologia Computacional , Dissulfetos/química , Biblioteca Gênica , Venenos de Escorpião/química , Sequência de Aminoácidos , Animais , Dados de Sequência Molecular , Escorpiões/metabolismo , Homologia de Sequência de Aminoácidos
18.
Toxicon ; 40(12): 1719-22, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12457884

RESUMO

A novel cDNA sequence encoding a new type of scorpion venom peptide (BmKAPi) was first isolated from the venom gland of Buthus martensii Karsch by cDNA library screening combined with 5'-race. The encoded precursor of BmKAPi consisted of 89 amino acid residues including a signal peptide of 24 residues, a putative mature peptide of 64 residues (BmKAPi) and an extra basic residue at the C-terminus which might be removed in the post-translational processing. BmKAPi is stabilized by five disulfide bridges, whereas all other disulfide-bridged scorpion toxins described are cross-linked by three or four disulfide bridges. It suggested the three-dimensional scaffold of BmKAPi might be different from other scorpion toxins. The amino acid sequence of BmKAPi showed no homology with other scorpion venom peptides, but shared a little similarity with some anticoagulant peptides and proteinase inhibitors isolated from hookworm, honeybee or European frog, respectively. RT-PCR analysis showed that BmKAPi mRNA could be induced by venom extraction suggesting BmKAPi might be a component of scorpion venom. These results suggest that BmKAPi is a new type of scorpion venom peptide different from other described scorpion toxins in structural and functional aspects.


Assuntos
Peptídeos/isolamento & purificação , Venenos de Escorpião/isolamento & purificação , Escorpiões/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Complementar , Dissulfetos/química , Biblioteca Gênica , Dados de Sequência Molecular , RNA Mensageiro , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Venenos de Escorpião/química , Venenos de Escorpião/genética , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA