Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 306(1): L50-7, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24213916

RESUMO

Alveolar type 2 (AT2) cells secrete surfactant that forms a protective layer on the lung's alveolar epithelium. Vesicles called lamellar bodies (LBs) store surfactant. Failure of surfactant secretion, which causes severe lung disease, relates to the manner in which LBs undergo exocytosis during the secretion. However, the dynamics of LBs during the secretion process are not known in intact alveoli. Here, we addressed this question through real-time confocal microscopy of single AT2 cells in live alveoli of mouse lungs. Using a combination of phospholipid and aqueous fluorophores that localize to LBs, we induced surfactant secretion by transiently hyperinflating the lung, and we quantified the secretion in terms of loss of bulk LB fluorescence. In addition, we quantified inter-LB phospholipid flow through determinations of fluorescence recovery after photobleaching. Furthermore, we determined the role of F-actin in surfactant secretion through expression of the fluorescent F-actin probe Lifeact. Our findings indicate that, in AT2 cells in situ, LBs are held in an F-actin scaffold. Although F-actin transiently decreases during surfactant secretion, the LBs remain stationary, forming a chain of vesicles connected by intervesicular channels that convey surfactant to the secretion site on the plasma membrane. This is the first instance of a secretory process in which the secretory vesicles are immobile, but form a conduit for the secretory material.


Assuntos
Actinas/metabolismo , Células Epiteliais Alveolares/metabolismo , Proteína B Associada a Surfactante Pulmonar/metabolismo , Vesículas Secretórias/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Camundongos , Multimerização Proteica , Ratos , Ratos Sprague-Dawley , Via Secretória , Técnicas de Cultura de Tecidos
2.
Am J Respir Cell Mol Biol ; 48(1): 78-86, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23043086

RESUMO

Although exposure to ambient hypoxia is known to cause proinflammatory vascular responses, the mechanisms initiating these responses are not understood. We tested the hypothesis that in systemic hypoxia, erythrocyte-derived H(2)O(2) induces proinflammatory gene transcription in vascular endothelium. We exposed mice or isolated, perfused murine lungs to 4 hours of hypoxia (8% O(2)). Leukocyte counts increased in the bronchoalveolar lavage. The expression of leukocyte adhesion receptors, reactive oxygen species, and protein tyrosine phosphorylation increased in freshly recovered lung endothelial cells (FLECs). These effects were inhibited by extracellular catalase and by the removal of erythrocytes, indicating that the responses were attributable to erythrocyte-derived H(2)O(2). Concomitant nuclear translocation of the p65 subunit of NF-κB and hypoxia-inducible factor-1α stabilization in FLECs occurred only in the presence of erythrocytes. Hemoglobin binding to the erythrocyte membrane protein, band 3, induced the release of H(2)O(2) from erythrocytes and the p65 translocation in FLECs. These data indicate for the first time, to our knowledge, that erythrocytes are responsible for endothelial transcriptional responses in hypoxia.


Assuntos
Eritrócitos/fisiologia , Hipóxia/sangue , Hipóxia/fisiopatologia , Animais , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Selectina E/sangue , Endotélio Vascular/fisiopatologia , Peróxido de Hidrogênio/sangue , Hipóxia/genética , Mediadores da Inflamação/sangue , Pulmão/irrigação sanguínea , Pulmão/fisiopatologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Transcrição/metabolismo
3.
Nat Commun ; 13(1): 5837, 2022 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-36192486

RESUMO

Acute Lung Injury (ALI) due to inhaled pathogens causes high mortality. Underlying mechanisms are inadequately understood. Here, by optical imaging of live mouse lungs we show that a key mechanism is the viability of cytosolic Ca2+ buffering by the mitochondrial Ca2+ uniporter (MCU) in the lung's surfactant-secreting, alveolar type 2 cells (AT2). The buffering increased mitochondrial Ca2+ and induced surfactant secretion in wild-type mice, but not in mice with AT2-specific MCU knockout. In the knockout mice, ALI due to intranasal LPS instillation caused severe pulmonary edema and mortality, which were mitigated by surfactant replenishment prior to LPS instillation, indicating surfactant's protective effect against alveolar edema. In wild-type mice, intranasal LPS, or Pseudomonas aeruginosa decreased AT2 MCU. Loss of MCU abrogated buffering. The resulting mortality was reduced by spontaneous recovery of MCU expression, or by MCU replenishment. Enhancement of AT2 mitochondrial buffering, hence endogenous surfactant secretion, through MCU replenishment might be a therapy against ALI.


Assuntos
Lesão Pulmonar Aguda , Lipopolissacarídeos , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Cálcio/metabolismo , Canais de Cálcio , Lipopolissacarídeos/toxicidade , Pulmão/metabolismo , Camundongos , Camundongos Knockout , Tensoativos
4.
bioRxiv ; 2021 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-33469582

RESUMO

Acute lung immunity to inhaled pathogens elicits defensive pneumonitis that may convert to the Acute Respiratory Distress Syndrome (ARDS), causing high mortality. Mechanisms underlying the conversion are not understood, but are of intense interest because of the ARDS-induced mortality in the ongoing Covid-19 pandemic. Here, by optical imaging of live lungs we show that key to the lethality is the functional status of mitochondrial Ca2+ buffering across the mitochondrial Ca2+ uniporter (MCU) in the alveolar type 2 cells (AT2), which protect alveolar stability. In mice subjected to ARDS by airway exposure to lipopolysaccharide (LPS), or to Pseudomonas aeruginosa, there was marked loss of MCU expression in AT2. The ability of mice to survive ARDS depended on the extent to which the MCU expression recovered, indicating that the viability of Ca2+ buffering by AT2 mitochondria critically determines ARDS severity. Mitochondrial transfer to enhance AT2 MCU expression might protect against ARDS.

5.
JCI Insight ; 6(6)2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33749665

RESUMO

High mortality in acute lung injury (ALI) results from sustained proinflammatory signaling by alveolar receptors, such as TNF-α receptor type 1 (TNFR1). Factors that determine the sustained signaling are not known. Unexpectedly, optical imaging of live alveoli revealed a major TNF-α-induced surge of alveolar TNFR1 due to a Ca2+-dependent mechanism that decreased the cortical actin fence. Mouse mortality due to inhaled LPS was associated with cofilin activation, actin loss, and the TNFR1 surge. The constitutively active form of the GTPase, Rac1 (V12Rac1), given intranasally (i.n.) as a noncovalent construct with a cell-permeable peptide, enhanced alveolar filamentous actin (F-actin) and blocked the TNFR1 surge. V12Rac1 also protected against ALI-induced mortality resulting from i.n. instillation of LPS or of Pseudomonas aeruginosa. We propose a potentially new therapeutic paradigm in which actin enhancement by exogenous Rac1 strengthens the alveolar actin fence, protecting against proinflammatory receptor hyperexpression, and therefore blocking ALI.


Assuntos
Actinas/uso terapêutico , Lesão Pulmonar Aguda/prevenção & controle , Neuropeptídeos/uso terapêutico , Proteínas rac1 de Ligação ao GTP/uso terapêutico , Lesão Pulmonar Aguda/metabolismo , Animais , Humanos , Masculino , Camundongos , Microscopia Confocal , Alvéolos Pulmonares/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo
6.
Nat Med ; 18(5): 759-65, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22504485

RESUMO

Bone marrow-derived stromal cells (BMSCs) protect against acute lung injury (ALI). To determine the role of BMSC mitochondria in this protection, we airway-instilled mice first with lipopolysaccharide (LPS) and then with either mouse BMSCs (mBMSCs) or human BMSCs (hBMSCs). Live optical studies revealed that the mBMSCs formed connexin 43 (Cx43)-containing gap junctional channels (GJCs) with the alveolar epithelia in these mice, releasing mitochondria-containing microvesicles that the epithelia engulfed. The presence of BMSC-derived mitochondria in the epithelia was evident optically, as well as by the presence of human mitochondrial DNA in mouse lungs instilled with hBMSCs. The mitochondrial transfer resulted in increased alveolar ATP concentrations. LPS-induced ALI, as indicated by alveolar leukocytosis and protein leak, inhibition of surfactant secretion and high mortality, was markedly abrogated by the instillation of wild-type mBMSCs but not of mutant, GJC-incompetent mBMSCs or mBMSCs with dysfunctional mitochondria. This is the first evidence, to our knowledge, that BMSCs protect against ALI by restituting alveolar bioenergetics through Cx43-dependent alveolar attachment and mitochondrial transfer.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Células da Medula Óssea/fisiologia , Mitocôndrias/fisiologia , Alvéolos Pulmonares/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Conexina 43/fisiologia , Metabolismo Energético , Junções Comunicantes/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Estromais/fisiologia
7.
J Clin Invest ; 121(5): 1986-99, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21519143

RESUMO

Shedding of the extracellular domain of cytokine receptors allows the diffusion of soluble receptors into the extracellular space; these then bind and neutralize their cytokine ligands, thus dampening inflammatory responses. The molecular mechanisms that control this process, and the extent to which shedding regulates cytokine-induced microvascular inflammation, are not well defined. Here, we used real-time confocal microscopy of mouse lung microvascular endothelium to demonstrate that mitochondria are key regulators of this process. The proinflammatory cytokine soluble TNF-α (sTNF-α) increased mitochondrial Ca2+, and the purinergic receptor P2Y2 prolonged the response. Concomitantly, the proinflammatory receptor TNF-α receptor-1 (TNFR1) was shed from the endothelial surface. Inhibiting the mitochondrial Ca2+ increase blocked the shedding and augmented inflammation, as denoted by increases in endothelial expression of the leukocyte adhesion receptor E-selectin and in microvascular leukocyte recruitment. The shedding was also blocked in microvessels after knockdown of a complex III component and after mitochondria-targeted catalase overexpression. Endothelial deletion of the TNF-α converting enzyme (TACE) prevented the TNF-α receptor shedding response, which suggests that exposure of microvascular endothelium to sTNF-α induced a Ca2+-dependent increase of mitochondrial H2O2 that caused TNFR1 shedding through TACE activation. These findings provide what we believe to be the first evidence that endothelial mitochondria regulate TNFR1 shedding and thereby determine the severity of sTNF-α-induced microvascular inflammation.


Assuntos
Cálcio/química , Pulmão/irrigação sanguínea , Mitocôndrias/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animais , Cálcio/metabolismo , Selectina E/metabolismo , Humanos , Peróxido de Hidrogênio/química , Inflamação , Leucócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação , Modelos Biológicos , Estrutura Terciária de Proteína , Espécies Reativas de Oxigênio , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA