Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 5837, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36192486

ABSTRACT

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.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Acute Lung Injury/chemically induced , Animals , Calcium/metabolism , Calcium Channels , Lipopolysaccharides/toxicity , Lung/metabolism , Mice , Mice, Knockout , Surface-Active Agents
2.
JCI Insight ; 6(6)2021 03 22.
Article in English | MEDLINE | ID: mdl-33749665

ABSTRACT

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.


Subject(s)
Actins/therapeutic use , Acute Lung Injury/prevention & control , Neuropeptides/therapeutic use , rac1 GTP-Binding Protein/therapeutic use , Acute Lung Injury/metabolism , Animals , Humans , Male , Mice , Microscopy, Confocal , Pulmonary Alveoli/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism
3.
bioRxiv ; 2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33469582

ABSTRACT

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.

4.
JCI Insight ; 4(3)2019 Feb 07.
Article in English | MEDLINE | ID: mdl-30728333

ABSTRACT

Acid aspiration, which can result from several etiologies, including postoperative complications, leads to direct contact of concentrated hydrochloric acid (HCl) with the alveolar epithelium. As a result, rapid endothelial activation induces alveolar inflammation, leading to life-threatening pulmonary edema. Because mechanisms underlying the rapid endothelial activation are not understood, here we determined responses in real time through optical imaging of alveoli of live mouse lungs. By alveolar micropuncture, we microinfused concentrated HCl in the alveolar lumen. As expected, acid contact with the epithelium caused rapid, but transient, apical injury. However, there was no concomitant membrane injury to the endothelium. Nevertheless, H2O2-mediated epithelial-endothelial paracrine signaling induced endothelial barrier failure, as detected by microvascular dextran leakage and lung water quantification. Remarkably, endothelial mitochondria regulated the barrier failure by activating uncoupling protein 2 (UCP2), thereby inducing transient mitochondrial depolarization that led to cofilin-induced actin depolymerization. Knockdown, or endothelium-targeted deletion of UCP2 expression, blocked these responses, including pulmonary edema. To our knowledge, these findings are the first to mechanistically implicate endothelial mitochondria in acid-induced barrier deterioration and pulmonary edema. We suggest endothelial UCP2 may be a therapeutic target for acid-induced acute lung injury.

5.
Am J Physiol Lung Cell Mol Physiol ; 306(1): L50-7, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24213916

ABSTRACT

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.


Subject(s)
Actins/metabolism , Alveolar Epithelial Cells/metabolism , Pulmonary Surfactant-Associated Protein B/metabolism , Secretory Vesicles/metabolism , Actin Cytoskeleton/metabolism , Animals , Mice , Protein Multimerization , Rats , Rats, Sprague-Dawley , Secretory Pathway , Tissue Culture Techniques
6.
Am J Respir Cell Mol Biol ; 48(1): 78-86, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23043086

ABSTRACT

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.


Subject(s)
Erythrocytes/physiology , Hypoxia/blood , Hypoxia/physiopathology , Animals , Anion Exchange Protein 1, Erythrocyte/metabolism , Bronchoalveolar Lavage Fluid/cytology , E-Selectin/blood , Endothelium, Vascular/physiopathology , Hydrogen Peroxide/blood , Hypoxia/genetics , Inflammation Mediators/blood , Lung/blood supply , Lung/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Transcription Factors/metabolism
7.
Am J Physiol Lung Cell Mol Physiol ; 302(11): L1209-20, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22505671

ABSTRACT

Although the lung expresses procoagulant proteins under inflammatory conditions, underlying mechanisms remain unclear. Here, we addressed lung endothelial expression of tissue factor (TF), which initiates the coagulation cascade and expression of which signifies development of a procoagulant phenotype in the vasculature. To establish the model of acid-induced acute lung injury (ALI), we intranasally instilled anesthetized mice with saline or acid. Then 2 h later, we isolated pulmonary vascular cells for flow cytometry and confocal microscopy to detect the leukocyte antigen, CD45 and the endothelial markers VE-cadherin and von Willebrand factor (vWf). Acid increased both the number of vWf-expressing cells as well as TF and P-selectin expressions on these cells. All of these effects were markedly inhibited by treating mice with antiplatelet serum, suggesting the involvement of platelets. The increased expressions of TF, vWf, and P-selectin in response to acid also occurred in platelets. Moreover, the effects were replicated in endothelial cells derived from isolated, blood-perfused lungs. However, the effect was inhibited completely in lungs perfused with platelet-depleted and, to a lesser extent, with leukocyte-depleted blood. Acid injury increased endothelial expressions of the platelet proteins, CD41 and CD42b, providing evidence that platelet proteins were transferred to the vascular surface. Reactive oxygen species (ROS) were implicated in these responses, in that the endothelial and platelet protein expressions were inhibited. We conclude that acid-induced ALI causes NOX2-mediated ROS generation that activates platelets, which then generate a procoagulant endothelial surface.


Subject(s)
Acute Lung Injury/blood , Acute Lung Injury/chemically induced , Blood Platelets/metabolism , Endothelium, Vascular/metabolism , Reactive Oxygen Species/metabolism , Thromboplastin/biosynthesis , Animals , Antigens, CD/biosynthesis , Blood Coagulation , Blood Platelets/immunology , Cadherins/biosynthesis , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hydrochloric Acid/adverse effects , Hydrochloric Acid/toxicity , Leukocyte Common Antigens/biosynthesis , Lung/immunology , Lung/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , P-Selectin/biosynthesis , P-Selectin/metabolism , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/immunology , Platelet Glycoprotein GPIb-IX Complex/biosynthesis , Platelet Membrane Glycoprotein IIb/biosynthesis , Thromboplastin/metabolism , von Willebrand Factor/biosynthesis , von Willebrand Factor/metabolism
8.
Nat Med ; 18(5): 759-65, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22504485

ABSTRACT

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.


Subject(s)
Acute Lung Injury/prevention & control , Bone Marrow Cells/physiology , Mitochondria/physiology , Pulmonary Alveoli/metabolism , Adenosine Triphosphate/metabolism , Animals , Connexin 43/physiology , Energy Metabolism , Gap Junctions/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Stromal Cells/physiology
9.
J Clin Invest ; 121(5): 1986-99, 2011 May.
Article in English | MEDLINE | ID: mdl-21519143

ABSTRACT

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.


Subject(s)
Calcium/chemistry , Lung/blood supply , Mitochondria/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Calcium/metabolism , E-Selectin/metabolism , Humans , Hydrogen Peroxide/chemistry , Inflammation , Leukocytes/cytology , Mice , Mice, Inbred C57BL , Microcirculation , Models, Biological , Protein Structure, Tertiary , Reactive Oxygen Species , Tumor Necrosis Factor-alpha/metabolism
10.
Blood ; 113(18): 4197-205, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19036701

ABSTRACT

Mesenchymal stem cells (MSCs), which potentially transdifferentiate into multiple cell types, are increasingly reported to be beneficial in models of organ system injury. However, the molecular mechanisms underlying interactions between MSCs and host cells, in particular endothelial cells (ECs), remain unclear. We show here in a matrigel angiogenesis assay that MSCs are capable of inhibiting capillary growth. After addition of MSCs to EC-derived capillaries in matrigel at EC:MSC ratio of 1:1, MSCs migrated toward the capillaries, intercalated between ECs, established Cx43-based intercellular gap junctional communication (GJC) with ECs, and increased production of reactive oxygen species (ROS). These events led to EC apoptosis and capillary degeneration. In an in vivo tumor model, direct MSC inoculation into subcutaneous melanomas induced apoptosis and abrogated tumor growth. Thus, our findings show for the first time that at high numbers, MSCs are potentially cytotoxic and that when injected locally in tumor tissue they might be effective antiangiogenesis agents suitable for cancer therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Endothelium, Vascular/cytology , Melanoma, Experimental/blood supply , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Neovascularization, Pathologic/therapy , Animals , Apoptosis/physiology , Cell Communication , Cells, Cultured , Collagen/metabolism , Drug Combinations , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Gap Junctions/physiology , Immunoblotting , Immunoenzyme Techniques , Immunoprecipitation , Laminin/metabolism , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Tissue Engineering
11.
Neuropharmacology ; 49(6): 715-23, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16129457

ABSTRACT

The extracellular concentration of the neurotransmitters dopamine, serotonin, norepinephrine, GABA and glycine is tightly controlled by plasma membrane transporters belonging to the SLC6 gene family. A very large number of putative transport proteins with a remarkable homology to the SLC6 transporters has recently been identified in prokaryotes. Here we have probed structural relationships in a 'microdoman' corresponding to the extracellular ends of transmembrane segments (TM) 7 and 8 in one of these homologs, the tryptophan transporter TnaT from Symbiobacterium thermophilum. We found that simultaneous - but not individual - substitution of Ala286 at the top of TM7 and Met311 at the top of TM8 with cysteines conferred sensitivity to submicromolar concentrations of Hg(2+) as assessed in a [(3)H]tryptophan uptake assay. Because Hg(2+) can cross-link pairs of cysteines, this suggests close proximity between TM 7 and 8 in the tertiary structure of TnaT as previously suggested for the mammalian counterparts. Furthermore, the inhibition of uptake upon cross-linking the two cysteines provides indirect support for a conserved conformational role of these transmembrane domains in the transport process. It was not possible, however, to transfer to TnaT binding sites for another metal ion, Zn(2+), that we previously engineered in the dopamine (DAT) and GABA (GAT-1) transporters between TM 7 and 8. This suggests that the structure of the TM7/8 microdomain is not identical with that of DAT and GAT-1. Hence, our data also emphasize possible structural differences that should be taken into account when interpreting future data on bacterial homologs of the SLC6 transporters.


Subject(s)
Biological Evolution , Membrane Transport Proteins/physiology , Protein Structure, Quaternary/physiology , Animals , Cross-Linking Reagents/pharmacology , Dose-Response Relationship, Drug , Escherichia coli , Extracellular Space , Gene Expression , Mammals , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Mercury/pharmacology , Mesylates/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding/drug effects , Sequence Homology, Amino Acid , Structure-Activity Relationship , Tritium/metabolism , Tryptophan/metabolism
12.
J Biol Chem ; 278(15): 12703-9, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12569103

ABSTRACT

The tnaT gene of Symbiobacterium thermophilum encodes a protein homologous to sodium-dependent neurotransmitter transporters. Expression of the tnaT gene product in Escherichia coli conferred the ability to accumulate tryptophan from the medium and the ability to grow on tryptophan as a sole source of carbon. Transport was Na(+)-dependent and highly selective. The K(m) for tryptophan was approximately 145 nm, and tryptophan transport was unchanged in the presence of 100 microM concentrations of other amino acids. Tryptamine and serotonin were weak inhibitors with K(I) values of 200 and 440 microM, respectively. By using a T7 promoter-based system, TnaT with an N-terminal His(6) tag was expressed at high levels in the membrane and was purified to near-homogeneity in high yield.


Subject(s)
Actinobacteria/metabolism , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins , Serotonin/metabolism , Tryptophan/metabolism , Actinobacteria/genetics , Amino Acid Sequence , Animals , Biological Transport/drug effects , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cloning, Molecular , Consensus Sequence , DNA Primers , Escherichia coli/genetics , Escherichia coli/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Rats , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Serotonin Plasma Membrane Transport Proteins , Sodium/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...