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

Bases de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Am J Respir Cell Mol Biol ; 71(1): 43-52, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38767348

RESUMO

Mechanical ventilation contributes to the morbidity and mortality of patients in intensive care, likely through the exacerbation and dissemination of inflammation. Despite the proximity of the pleural cavity to the lungs and exposure to physical forces, little attention has been paid to its potential as an inflammatory source during ventilation. Here, we investigate the pleural cavity as a novel site of inflammation during ventilator-induced lung injury. Mice were subjected to low or high tidal volume ventilation strategies for up to 3 hours. Ventilation with a high tidal volume significantly increased cytokine and total protein levels in BAL and pleural lavage fluid. In contrast, acid aspiration, explored as an alternative model of injury, only promoted intraalveolar inflammation, with no effect on the pleural space. Resident pleural macrophages demonstrated enhanced activation after injurious ventilation, including upregulated ICAM-1 and IL-1ß expression, and the release of extracellular vesicles. In vivo ventilation and in vitro stretch of pleural mesothelial cells promoted ATP secretion, whereas purinergic receptor inhibition substantially attenuated extracellular vesicles and cytokine levels in the pleural space. Finally, labeled protein rapidly translocated from the pleural cavity into the circulation during high tidal volume ventilation, to a significantly greater extent than that of protein translocation from the alveolar space. Overall, we conclude that injurious ventilation induces pleural cavity inflammation mediated through purinergic pathway signaling and likely enhances the dissemination of mediators into the vasculature. This previously unidentified consequence of mechanical ventilation potentially implicates the pleural space as a focus of research and novel avenue for intervention in critical care.


Assuntos
Camundongos Endogâmicos C57BL , Cavidade Pleural , Lesão Pulmonar Induzida por Ventilação Mecânica , Animais , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia , Cavidade Pleural/metabolismo , Cavidade Pleural/patologia , Inflamação/patologia , Inflamação/metabolismo , Camundongos , Respiração Artificial/efeitos adversos , Volume de Ventilação Pulmonar , Macrófagos/metabolismo , Macrófagos/patologia , Trifosfato de Adenosina/metabolismo , Vesículas Extracelulares/metabolismo , Masculino , Citocinas/metabolismo , Líquido da Lavagem Broncoalveolar , Modelos Animais de Doenças , Interleucina-1beta/metabolismo
2.
Am J Respir Crit Care Med ; 204(4): 421-430, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-33848447

RESUMO

Rationale: Mechanical ventilation is a mainstay of intensive care but contributes to the mortality of patients through ventilator-induced lung injury. eCypA (extracellular CypA [cyclophilin A]) is an emerging inflammatory mediator and metalloproteinase inducer, and the gene responsible for its expression has recently been linked to coronavirus disease (COVID-19). Objectives: To explore the involvement of eCypA in the pathophysiology of ventilator-induced lung injury. Methods: Mice were ventilated with a low or high Vt for up to 3 hours, with or without blockade of eCypA signaling, and lung injury and inflammation were evaluated. Human primary alveolar epithelial cells were exposed to in vitro stretching to explore the cellular source of eCypA, and CypA concentrations were measured in BAL fluid from patients with acute respiratory distress syndrome to evaluate the clinical relevance. Measurements and Main Results: High-Vt ventilation in mice provoked a rapid increase in soluble CypA concentration in the alveolar space but not in plasma. In vivo ventilation and in vitro stretching experiments indicated the alveolar epithelium as the likely major source. In vivo blockade of eCypA signaling substantially attenuated physiological dysfunction, macrophage activation, and MMPs (matrix metalloproteinases). Finally, we found that patients with acute respiratory distress syndrome showed markedly elevated concentrations of eCypA within BAL fluid. Conclusions: CypA is upregulated within the lungs of injuriously ventilated mice (and critically ill patients), where it plays a significant role in lung injury. eCypA represents an exciting novel target for pharmacological intervention.


Assuntos
Anti-Inflamatórios/imunologia , Ciclofilina A/imunologia , Inflamação/imunologia , Respiração Artificial/efeitos adversos , Síndrome do Desconforto Respiratório/imunologia , Mucosa Respiratória/imunologia , Lesão Pulmonar Induzida por Ventilação Mecânica/imunologia , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia , Animais , COVID-19/genética , COVID-19/fisiopatologia , Células Cultivadas/efeitos dos fármacos , Ciclofilina A/farmacologia , Humanos , Inflamação/fisiopatologia , Masculino , Camundongos , Modelos Animais , Síndrome do Desconforto Respiratório/fisiopatologia , SARS-CoV-2 , Lesão Pulmonar Induzida por Ventilação Mecânica/genética
3.
Am J Physiol Lung Cell Mol Physiol ; 320(1): L73-L83, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33146567

RESUMO

Despite advances in the pathophysiology of chronic obstructive pulmonary disease (COPD), there is a distinct lack of biochemical markers to aid clinical management. Microvesicles (MVs) have been implicated in the pathophysiology of inflammatory diseases including COPD, but their association to COPD disease severity remains unknown. We analyzed different MV populations in plasma and bronchoalveolar lavage fluid (BALF) taken from 62 patients with mild to very severe COPD (51% male; mean age: 65.9 yr). These patients underwent comprehensive clinical evaluation (symptom scores, lung function, and exercise testing), and the capacity of MVs to be clinical markers of disease severity was assessed. We successfully identified various MV subtype populations within BALF [leukocyte, polymorphonuclear leukocyte (PMN; i.e., neutrophil), monocyte, epithelial, and platelet MVs] and plasma (leukocyte, PMN, monocyte, and endothelial MVs) and compared each MV population to disease severity. BALF neutrophil MVs were the only population to significantly correlate with the clinical evaluation scores including forced expiratory volume in 1 s, modified Medical Research Council dyspnea score, 6-min walk test, hyperinflation, and gas transfer. BALF neutrophil MVs, but not neutrophil cell numbers, also strongly correlated with BODE index. We have undertaken, for the first time, a comprehensive evaluation of MV profiles within BALF/plasma of COPD patients. We demonstrate that BALF levels of neutrophil-derived MVs are unique in correlating with a number of key functional and clinically relevant disease severity indexes. Our results show the potential of BALF neutrophil MVs for a COPD biomarker that tightly links a key pathophysiological mechanism of COPD (intra-alveolar neutrophil activation) with clinical severity/outcome.


Assuntos
Líquido da Lavagem Broncoalveolar/química , Micropartículas Derivadas de Células/patologia , Neutrófilos/patologia , Alvéolos Pulmonares/patologia , Doença Pulmonar Obstrutiva Crônica/patologia , Índice de Gravidade de Doença , Idoso , Micropartículas Derivadas de Células/metabolismo , Citocinas/metabolismo , Feminino , Volume Expiratório Forçado , Humanos , Masculino , Neutrófilos/metabolismo , Alvéolos Pulmonares/metabolismo , Doença Pulmonar Obstrutiva Crônica/etiologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Testes de Função Respiratória
4.
FASEB J ; 33(5): 6442-6455, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30776316

RESUMO

Cellular stress or injury induces release of endogenous danger signals such as ATP, which plays a central role in activating immune cells. ATP is essential for the release of nonclassically secreted cytokines such as IL-1ß but, paradoxically, has been reported to inhibit the release of classically secreted cytokines such as TNF. Here, we reveal that ATP does switch off soluble TNF (17 kDa) release from LPS-treated macrophages, but rather than inhibiting the entire TNF secretion, ATP packages membrane TNF (26 kDa) within microvesicles (MVs). Secretion of membrane TNF within MVs bypasses the conventional endoplasmic reticulum- and Golgi transport-dependent pathway and is mediated by acid sphingomyelinase. These membrane TNF-carrying MVs are biologically more potent than soluble TNF in vivo, producing significant lung inflammation in mice. Thus, ATP critically alters TNF trafficking and secretion from macrophages, inducing novel unconventional membrane TNF signaling via MVs without direct cell-to-cell contact. These data have crucial implications for this key cytokine, particularly when therapeutically targeting TNF in acute inflammatory diseases.-Soni, S., O'Dea, K. P., Tan, Y. Y., Cho, K., Abe, E., Romano, R., Cui, J., Ma, D., Sarathchandra, P., Wilson, M. R., Takata, M. ATP redirects cytokine trafficking and promotes novel membrane TNF signaling via microvesicles.


Assuntos
Trifosfato de Adenosina/imunologia , Membrana Celular/imunologia , Vesículas Extracelulares/imunologia , Macrófagos/imunologia , Pneumonia/imunologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/imunologia , Doença Aguda , Trifosfato de Adenosina/genética , Animais , Comunicação Celular/genética , Comunicação Celular/imunologia , Membrana Celular/genética , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Vesículas Extracelulares/genética , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Knockout , Pneumonia/induzido quimicamente , Pneumonia/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Necrose Tumoral alfa/genética
5.
Thorax ; 74(12): 1120-1129, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31278170

RESUMO

BACKGROUND: Despite advances in understanding the pathophysiology of acute respiratory distress syndrome, effective pharmacological interventions have proven elusive. We believe this is a consequence of existing preclinical models being designed primarily to explore biological pathways, rather than predict treatment effects. Here, we describe a mouse model in which both therapeutic intervention and ventilation were superimposed onto existing injury and explored the impact of ß-agonist treatment, which is effective in simple models but not clinically. METHODS: Mice had lung injury induced by intranasal lipopolysaccharide (LPS), which peaked at 48 hours post-LPS based on clinically relevant parameters including hypoxaemia and impaired mechanics. At this peak of injury, mice were treated intratracheally with either terbutaline or tumour necrosis factor (TNF) receptor 1-targeting domain antibody, and ventilated with moderate tidal volume (20 mL/kg) to induce secondary ventilator-induced lung injury (VILI). RESULTS: Ventilation of LPS-injured mice at 20 mL/kg exacerbated injury compared with low tidal volume (8 mL/kg). While terbutaline attenuated VILI within non-LPS-treated animals, it was ineffective to reduce VILI in pre-injured mice, mimicking its lack of clinical efficacy. In contrast, anti-TNF receptor 1 antibody attenuated secondary VILI within pre-injured lungs, indicating that the model was treatable. CONCLUSIONS: We propose adoption of a practical framework like that described here to reduce the number of ultimately ineffective drugs reaching clinical trials. Novel targets should be evaluated alongside interventions which have been previously tested clinically, using models that recapitulate the (lack of) clinical efficacy. Within such a framework, outperforming a failed pharmacologic should be a prerequisite for drugs entering trials.


Assuntos
Respiração Artificial/efeitos adversos , Síndrome do Desconforto Respiratório/terapia , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Agonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Animais , Anticorpos Neutralizantes/uso terapêutico , Modelos Animais de Doenças , Lipopolissacarídeos , Masculino , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Respiração Artificial/métodos , Síndrome do Desconforto Respiratório/induzido quimicamente , Síndrome do Desconforto Respiratório/fisiopatologia , Terbutalina/uso terapêutico , Volume de Ventilação Pulmonar , Lesão Pulmonar Induzida por Ventilação Mecânica/etiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia
10.
Thorax ; 71(11): 1020-1029, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27287089

RESUMO

BACKGROUND: Microvesicles (MVs) are important mediators of intercellular communication, packaging a variety of molecular cargo. They have been implicated in the pathophysiology of various inflammatory diseases; yet, their role in acute lung injury (ALI) remains unknown. OBJECTIVES: We aimed to identify the biological activity and functional role of intra-alveolar MVs in ALI. METHODS: Lipopolysaccharide (LPS) was instilled intratracheally into C57BL/6 mice, and MV populations in bronchoalveolar lavage fluid (BALF) were evaluated. BALF MVs were isolated 1 hour post LPS, assessed for cytokine content and incubated with murine lung epithelial (MLE-12) cells. In separate experiments, primary alveolar macrophage-derived MVs were incubated with MLE-12 cells or instilled intratracheally into mice. RESULTS: Alveolar macrophages and epithelial cells rapidly released MVs into the alveoli following LPS. At 1 hour, the dominant population was alveolar macrophage-derived, and these MVs carried substantive amounts of tumour necrosis factor (TNF) but minimal amounts of IL-1ß/IL-6. Incubation of these mixed MVs with MLE-12 cells induced epithelial intercellular adhesion molecule-1 (ICAM-1) expression and keratinocyte-derived cytokine release compared with MVs from untreated mice (p<0.001). MVs released in vitro from LPS-primed alveolar macrophages caused similar increases in MLE-12 ICAM-1 expression, which was mediated by TNF. When instilled intratracheally into mice, these MVs induced increases in BALF neutrophils, protein and epithelial cell ICAM-1 expression (p<0.05). CONCLUSIONS: We demonstrate, for the first time, the sequential production of MVs from different intra-alveolar precursor cells during the early phase of ALI. Our findings suggest that alveolar macrophage-derived MVs, which carry biologically active TNF, may play an important role in initiating ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/fisiopatologia , Micropartículas Derivadas de Células/metabolismo , Macrófagos Alveolares/metabolismo , Animais , Líquido da Lavagem Broncoalveolar/citologia , Citocinas/metabolismo , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL
13.
Front Immunol ; 13: 853769, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35572508

RESUMO

Intra-alveolar microvesicles (MVs) are important mediators of inter-cellular communication within the alveolar space, and are key components in the pathophysiology of lung inflammation such as acute respiratory distress syndrome (ARDS). Despite the abundance of data detailing the pro-inflammatory effects of MVs, it remains unclear how MVs interact or signal with target cells in the alveolus. Using both in vivo and in vitro alveolar models, we analyzed the dynamics of MV uptake by resident alveolar cells: alveolar macrophages and epithelial cells. Under resting conditions, the overwhelming majority of MVs were taken up by alveolar macrophages. However, following lipopolysaccharide (LPS)-mediated inflammation, epithelial cells internalized significantly more MVs (p<0.01) whilst alveolar macrophage internalization was significantly reduced (p<0.01). We found that alveolar macrophages adopted a pro-inflammatory phenotype after internalizing MVs under resting conditions, but reduction of MV uptake following LPS pre-treatment was associated with loss of inflammatory phenotype. Instead, MVs induced significant epithelial cell inflammation following LPS pre-treatment, when MV internalization was most significant. Using pharmacological inhibitors, we interrogated the mechanisms of MV internalization to identify which endocytic pathways and cell surface receptors are involved. We demonstrated that epithelial cells are exclusively dependent on the clathrin and caveolin dependent endocytotic pathway, whereas alveolar macrophage uptake may involve a significant phagocytic component. Furthermore, alveolar macrophages predominantly engulf MVs via scavenger receptors whilst, epithelial cells internalize MVs via a phosphatidylserine/integrin receptor mediated pathway (specifically alpha V beta III), which can be inhibited with phosphatidylserine-binding protein (i.e. annexin V). In summary, we have undertaken a comprehensive evaluation of MV internalization within the alveolar space. Our results demonstrate that different environmental conditions can modulate MV internalization, with inflammatory stimuli strongly enhancing epithelial cell uptake of MVs and inducing epithelial cell activation. Our data reveal the unique mechanisms by which alveolar macrophages and epithelial cells internalize MVs thereby elucidating how MVs exert their pathophysiological effect during lung inflammation and injury. As MVs are potential novel therapeutic targets in conditions such as ARDS, these data provide crucial insights into the dynamics of MV-target cell interactions and highlight potential avenues for researchers to modulate and inhibit their pro-inflammatory actions within the alveolar space.


Assuntos
Pneumonia , Síndrome do Desconforto Respiratório , Células Epiteliais , Humanos , Inflamação/metabolismo , Lipopolissacarídeos/metabolismo , Macrófagos Alveolares/metabolismo , Fosfatidilserinas/metabolismo , Pneumonia/metabolismo
14.
PLoS One ; 17(7): e0269244, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35776718

RESUMO

A number of studies have highlighted physiological data from the first surge in critically unwell Covid-19 patients but there is a paucity of data describing emerging variants of SARS-CoV-2, such as B.1.1.7. We compared ventilatory parameters, biochemical and physiological data and mortality between the first and second COVID-19 surges in the United Kingdom, where distinct variants of SARS-CoV-2 were the dominant stain. We performed a retrospective cohort study investigating critically unwell patients admitted with COVID-19 across three tertiary regional ICUs in London, UK. Of 1782 adult ICU patients screened, 330 intubated and ventilated patients diagnosed with COVID-19 were included. In the second wave where B.1.1.7 variant was the dominant strain, patients were had increased severity of ARDS whilst compliance was greater (p<0.05) and d-dimer lower. The 28-day mortality was not statistically significant (1st wave: 42.2% vs 2nd wave: 39.8%). However, when adjusted for key covariates, the hazard ratio for 28-day mortality in those patients with B.1.1.7 was 3.79 (CI 1.04-13.8; p = 0.043) compared to the original strain. During the second surge in the UK, where the COVID-19 variant B.1.1.7 was most prevalent, significantly more patients presented to critical care with severe ARDS. Furthermore, mortality risk was significantly greater in our ICU population during the second wave of the pandemic in those patients with B.1.1.7. As ICUs are experiencing further waves (particularly by the delta (B.1.617.2) variant), we highlight the urgent need for prospective studies describing immunological and pathophysiological differences across novel emerging variants.


Assuntos
COVID-19 , Síndrome do Desconforto Respiratório , Adulto , Cuidados Críticos , Humanos , Estudos Prospectivos , Estudos Retrospectivos , SARS-CoV-2
15.
BMJ Case Rep ; 14(3)2021 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-33782066

RESUMO

Emerging data suggest that patients with certain comorbidities requiring intensive care unit (ICU) admission for COVID-19 have a poor prognosis. This report describes a case of a patient with multiple comorbidities who contracted COVID-19 pneumonitis but was successfully weaned off invasive mechanical ventilation after 118 days, despite his admission being complicated by recurrent septic episodes and requirement for advanced cardiovascular support and renal replacement therapy. Of note, our patient received three courses of steroids in total during his ICU stay,and current literature strongly supports the use of steroids in critically unwell patients with COVID-19. To the best of our knowledge, this is the longest reported ventilated time and intensive care/hospital stay for a surviving patient with COVID-19 and highlights the importance of allowing sufficient time for clinical interventions to take effect, even when the prognosis appears bleak.


Assuntos
COVID-19/terapia , Pneumonia/terapia , Respiração Artificial , Anti-Infecciosos/uso terapêutico , COVID-19/complicações , Comorbidade , Humanos , Masculino , Pessoa de Meia-Idade , Pneumonia/virologia , Terapia de Substituição Renal , Sepse/complicações , Sepse/tratamento farmacológico , Esteroides/uso terapêutico , Reino Unido , Desmame do Respirador , Ventiladores Mecânicos
16.
J Extracell Vesicles ; 9(1): 1706708, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32002170

RESUMO

Microvesicles (MVs), a plasma membrane-derived subclass of extracellular vesicles, are produced and released into the circulation during systemic inflammation, yet little is known of cell/tissue-specific uptake of MVs under these conditions. We hypothesized that monocytes contribute to uptake of circulating MVs and that their increased margination to the pulmonary circulation and functional priming during systemic inflammation produces substantive changes to the systemic MV homing profile. Cellular uptake of i.v.-injected, fluorescently labelled MVs (J774.1 macrophage-derived) in vivo was quantified by flow cytometry in vascular cell populations of the lungs, liver and spleen of C57BL6 mice. Under normal conditions, both Ly6Chigh and Ly6Clow monocytes contributed to MV uptake but liver Kupffer cells were the dominant target cell population. Following induction of sub-clinical endotoxemia with low-dose i.v. LPS, MV uptake by lung-marginated Ly6Chigh monocytes increased markedly, both at the individual cell level (~2.5-fold) and through substantive expansion of their numbers (~8-fold), whereas uptake by splenic macrophages was unchanged and uptake by Kupffer cells actually decreased (~50%). Further analysis of MV uptake within the pulmonary vasculature using a combined model approach of in vivo macrophage depletion, ex vivo isolated perfused lungs and in vitro lung perfusate cell-based assays, indicated that Ly6Chigh monocytes possess a high MV uptake capacity (equivalent to Kupffer cells), that is enhanced directly by endotoxemia and ablated in the presence of phosphatidylserine (PS)-enriched liposomes and ß3 integrin receptor blocking peptide. Accordingly, i.v.-injected PS-enriched liposomes underwent a redistribution of cellular uptake during endotoxemia similar to MVs, with enhanced uptake by Ly6Chigh monocytes and reduced uptake by Kupffer cells. These findings indicate that monocytes, particularly lung-marginated Ly6Chigh subset monocytes, become a dominant target cell population for MVs during systemic inflammation, with significant implications for the function and targeting of endogenous and therapeutically administered MVs, lending novel insights into the pathophysiology of pulmonary vascular inflammation.

17.
Expert Opin Ther Targets ; 23(11): 931-941, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31724440

RESUMO

Introduction: Acute respiratory distress syndrome (ARDS) is a heterogeneous and multifactorial disease; it is a common and devastating condition that has a high mortality. Treatment is limited to supportive measures hence novel pharmacological approaches are necessary. We propose a new direction in ARDS research; this means moving away from thinking about individual inflammatory mediators and instead investigating how packaged information is transmitted between cells. Microvesicles (MVs) represent a novel vehicle for inter-cellular communication with an emerging role in ARDS pathophysiology.Areas covered: This review examines current approaches to ARDS and emerging MV research. We describe advances in our understanding of microvesicles and focus on their pro-inflammatory roles in airway and endothelial signaling. We also offer reasons for why MVs are attractive therapeutic targets.Expert opinion: MVs have a key role in ARDS pathophysiology. Preclinical studies must move away from simple models toward more realistic scenarios while clinical studies must embrace patient heterogeneity. Microvesicles have the potential to aid identification of patients who may benefit from particular treatments and act as biomarkers of cellular status and disease progression. Understanding microvesicle cargoes and their cellular interactions will undoubtedly uncover new targets for ARDS.


Assuntos
Vesículas Extracelulares/metabolismo , Terapia de Alvo Molecular , Síndrome do Desconforto Respiratório/tratamento farmacológico , Animais , Comunicação Celular/fisiologia , Humanos , Modelos Biológicos , Síndrome do Desconforto Respiratório/fisiopatologia
19.
Int J Emerg Med ; 4: 8, 2011 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-21468363

RESUMO

Intussusception is an uncommon cause of abdominal pain in adults and poses diagnostic challenges for emergency physicians, due to its varied presenting symptoms and time course. Diagnosis is thus often delayed and results in surgical intervention due to the development of bowel ischaemia. We report on a young patient who presented with an ileo-ileal intussusception in whom there were no underlying lesions identified as a causal factor.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA