Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Hepatology ; 72(6): 2165-2181, 2020 12.
Article in English | MEDLINE | ID: mdl-32190913

ABSTRACT

BACKGROUND AND AIMS: Hepatic crisis is an emergent complication affecting patients with sickle cell disease (SCD); however, the molecular mechanism of sickle cell hepatobiliary injury remains poorly understood. Using the knock-in humanized mouse model of SCD and SCD patient blood, we sought to mechanistically characterize SCD-associated hepato-pathophysiology applying our recently developed quantitative liver intravital imaging, RNA sequence analysis, and biochemical approaches. APPROACH AND RESULTS: SCD mice manifested sinusoidal ischemia, progressive hepatomegaly, liver injury, hyperbilirubinemia, and increased ductular reaction under basal conditions. Nuclear factor kappa B (NF-κB) activation in the liver of SCD mice inhibited farnesoid X receptor (FXR) signaling and its downstream targets, leading to loss of canalicular bile transport and altered bile acid pool. Intravital imaging revealed impaired bile secretion into the bile canaliculi, which was secondary to loss of canalicular bile transport and bile acid metabolism, leading to intrahepatic bile accumulation in SCD mouse liver. Blocking NF-κB activation rescued FXR signaling and partially ameliorated liver injury and sinusoidal ischemia in SCD mice. CONCLUSIONS: These findings identify that NF-κB/FXR-dependent impaired bile secretion promotes intrahepatic bile accumulation, which contributes to hepatobiliary injury of SCD. Improved understanding of these processes could potentially benefit the development of therapies to treat sickle cell hepatic crisis.


Subject(s)
Anemia, Sickle Cell/complications , Bile/metabolism , Cholestasis/etiology , Hepatic Insufficiency/etiology , Liver/pathology , Adolescent , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Animals , Bile Ducts, Intrahepatic/diagnostic imaging , Bile Ducts, Intrahepatic/pathology , Cholestasis/pathology , Cholestasis/prevention & control , Disease Models, Animal , Female , Gene Knock-In Techniques , Hemoglobin, Sickle/genetics , Hepatic Insufficiency/pathology , Hepatic Insufficiency/prevention & control , Humans , Intravital Microscopy , Liver/diagnostic imaging , Male , Mice , Middle Aged , NF-kappa B/antagonists & inhibitors , NF-kappa B/drug effects , NF-kappa B/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Young Adult
2.
Respirology ; 25(4): 417-426, 2020 04.
Article in English | MEDLINE | ID: mdl-31364255

ABSTRACT

BACKGROUND AND OBJECTIVE: Mesenchymal stem cells (MSC) have been shown to ameliorate the deleterious effects of bleomycin in murine models. However, the mechanism responsible for protection from pulmonary fibrosis by stem cell therapy is still poorly understood, especially in terms of endoplasmic reticulum (ER) stress. We hypothesized that during bleomycin-induced lung injury, markers of ER stress, specifically the activation of the unfolded protein response (UPR), increase during injury, resembling the kinetics of collagen deposition in the lung described for the bleomycin model. We aimed to elucidate the possible role of MSC in ER stress modulation. METHODS: To determine the kinetics of ER stress in aged mice, the expression of ER stress markers after bleomycin lung injury was measured in old mice at different time points (days 0, 3, 7, 14 and 21). To evaluate the consequences of systemic delivery of MSC on lung ER stress in the bleomycin model, we evaluated changes in body weight, lung histology and protein expression of ER stress markers. RESULTS: The level of expression of UPR transcription factor XBP-1 and its regulator BiP was elevated at day 7 and progressively increased up to day 21. MSC inhibited BiP expression in bleomycin-induced ER stress, attenuating ER stress via the protein kinase RNA-like ER kinase (PERK)-Nrf2 pathway. The expression levels of other ER stress markers were not perturbed by MSC. CONCLUSION: Our data suggest that MSC operate on ER stress via several pathways, but the PERK-Nrf2 pathway revealed to be the main functioning pathway in our bleomycin model.


Subject(s)
Endoplasmic Reticulum Stress , Mesenchymal Stem Cell Transplantation , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/therapy , Unfolded Protein Response , Animals , Bleomycin , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/metabolism , Humans , Mice , NF-E2-Related Factor 2/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/physiopathology , X-Box Binding Protein 1/metabolism , eIF-2 Kinase/metabolism
3.
Am J Respir Cell Mol Biol ; 60(6): 629-636, 2019 06.
Article in English | MEDLINE | ID: mdl-30543447

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) pathogenesis has been postulated to involve a variety of mechanisms associated with the aging process, including loss of protein homeostasis (proteostasis). Heat shock proteins are cellular chaperones that serve a number of vital maintenance and repair functions, including the regulation of proteostasis. Previously published data have implicated heat shock protein 70 (Hsp70) in the development of pulmonary fibrosis in animal models. We sought to identify alterations in Hsp70 expression in IPF lung. Hsp70 mRNA and protein were decreased in primary fibroblasts cultured from IPF versus normal donor lung tissue. In addition to cultured fibroblasts, Hsp70 expression was decreased in intact IPF lung, a stressed environment in which upregulation of protective heat shock proteins would be anticipated. In support of a mechanistic association between decreased Hsp70 and fibrosis, cultured primary lung fibroblasts deficient in Hsp70 secreted increased extracellular matrix proteins. Treatment of primary normal human lung fibroblasts in vitro with either of the profibrotic molecules IGFBP5 (insulin-like growth factor-binding protein 5) or transforming growth factor-ß1 downregulated Hsp70, suggesting Hsp70 is a downstream target in the fibrotic cascade. Hsp70-knockout mice subjected to an inhalational bleomycin model of pulmonary fibrosis demonstrated accelerated fibrosis versus wild-type control animals. We therefore conclude that reduced Hsp70 protein contributes to fibrosis and that interventions aimed at restoring normal expression of Hsp70 represent a novel therapeutic strategy for pulmonary fibrosis.


Subject(s)
HSP70 Heat-Shock Proteins/deficiency , Idiopathic Pulmonary Fibrosis/metabolism , Intracellular Space/metabolism , Aging/pathology , Animals , Bleomycin , Fibroblasts/metabolism , Fibroblasts/pathology , HSP70 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 5/metabolism , Lung/pathology , Mice , Phenotype , Transforming Growth Factor beta1/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 317(4): L510-L521, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31432710

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is the most common and devastating of the interstitial lung diseases. Epithelial dysfunction is thought to play a prominent role in disease pathology, and we sought to characterize secreted signals that may contribute to disease pathology. Transcriptional profiling of senescent type II alveolar epithelial cells from mice with epithelial-specific telomere dysfunction identified the transforming growth factor-ß family member, growth and differentiation factor 15 (Gdf15), as the most significantly upregulated secreted protein. Gdf15 expression is induced in response to telomere dysfunction and bleomycin challenge in mice. Gdf15 mRNA is expressed by lung epithelial cells, and protein can be detected in peripheral blood and bronchoalveolar lavage following bleomycin challenge in mice. In patients with IPF, GDF15 mRNA expression in lung tissue is significantly increased and correlates with pulmonary function. Single-cell RNA sequencing of human lungs identifies epithelial cells as the primary source of GDF15, and circulating concentrations of GDF15 are markedly elevated and correlate with disease severity and survival in multiple independent cohorts. Our findings suggest that GDF15 is an epithelial-derived secreted protein that may be a useful biomarker of epithelial stress and identifies IPF patients with poor outcomes.


Subject(s)
Alveolar Epithelial Cells/metabolism , Growth Differentiation Factor 15/genetics , Idiopathic Pulmonary Fibrosis/genetics , Transcriptome , Aged , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology , Animals , Bleomycin/administration & dosage , Bronchoalveolar Lavage Fluid/chemistry , Case-Control Studies , Disease Models, Animal , Female , Gene Expression Profiling , Growth Differentiation Factor 15/metabolism , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/mortality , Idiopathic Pulmonary Fibrosis/physiopathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Middle Aged , Respiratory Function Tests , Severity of Illness Index , Survival Analysis , Telomere
5.
Am J Physiol Lung Cell Mol Physiol ; 313(1): L92-L103, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28385811

ABSTRACT

Although different preclinical models have demonstrated a favorable role for bone marrow-derived mesenchymal stem cells (B-MSC) in preventing fibrosis, this protective effect is not observed with late administration of these cells, when fibrotic changes are consolidated. We sought to investigate whether the late administration of B-MSCs overexpressing microRNAs (miRNAs) let-7d (antifibrotic) or miR-154 (profibrotic) could alter lung fibrosis in a murine bleomycin model. Using lentiviral vectors, we transduced miRNAs (let-7d or miR-154) or a control sequence into human B-MSCs. Overexpression of let-7d or miR-154 was associated with changes in the mesenchymal properties of B-MSCs and in their cytokine expression. Modified B-MSCs were intravenously administered to mice at day 7 after bleomycin instillation, and the mice were euthanized at day 14 Bleomycin-injured animals that were treated with let-7d cells were found to recover quicker from the initial weight loss compared with the other treatment groups. Interestingly, animals treated with miR-154 cells had the lowest survival rate. Although a slight reduction in collagen mRNA levels was observed in lung tissue from let-7d mice, no significant differences were observed in Ashcroft score and OH-proline. However, the distinctive expression in cytokines and CD45-positive cells in the lung suggests that the differential effects observed in both miRNA mice groups were related to an effect on the immunomodulation function. Our results establish the use of miRNA-modified mesenchymal stem cells as a potential future research in lung fibrosis.


Subject(s)
Lung Injury/metabolism , Lung Injury/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Transduction, Genetic , Animals , Biomarkers/metabolism , Bleomycin , Bone Marrow Cells/cytology , Collagen/genetics , Collagen/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Gene Regulatory Networks , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Leukocyte Common Antigens/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , Transfection , Weight Loss
6.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L1164-L1173, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28860144

ABSTRACT

The mechanisms of aging that are involved in the development of idiopathic pulmonary fibrosis (IPF) are still unclear. Although it has been hypothesized that the proliferation and activation of human lung fibroblasts (hLFs) are essential in IPF, no studies have assessed how this process works in an aging lung. Our goal was to elucidate if there were age-related changes on primary hLFs isolated from IPF lungs compared with age-matched controls. We investigated several hallmarks of aging in hLFs from IPF patients and age-matched controls. IPF hLFs have increased cellular senescence with higher expression of ß-galactosidase, p21, p16, p53, and cytokines related to the senescence-associated secretory phenotype (SASP) as well as decreased proliferation/apoptosis compared with age-matched controls. Additionally, we observed shorter telomeres, mitochondrial dysfunction, and upon transforming growth factor-ß stimulation, increased markers of endoplasmic reticulum stress. Our data suggest that IPF hLFs develop senescence resulting in a decreased apoptosis and that the development of SASP may be an important contributor to the fibrotic process observed in IPF. These results might change the existing paradigm, which describes fibroblasts as aberrantly activated cells, to a cell with a senescence phenotype.


Subject(s)
Aging/metabolism , Cellular Senescence , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Adult , Aging/pathology , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytokines/metabolism , Female , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Male , Middle Aged , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/metabolism
7.
Blood ; 123(18): 2864-72, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24677541

ABSTRACT

Bioenergetic dysfunction, although central to the pathogenesis of numerous diseases, remains uncharacterized in many patient populations because of the invasiveness of obtaining tissue for mitochondrial studies. Although platelets are an accessible source of mitochondria, the role of bioenergetics in regulating platelet function remains unclear. Herein, we validate extracellular flux analysis in human platelets and use this technique to screen for mitochondrial dysfunction in sickle cell disease (SCD) patients, a population with aberrant platelet activation of an unknown mechanism and in which mitochondrial function has never been assessed. We identify a bioenergetic alteration in SCD patients characterized by deficient complex V activity, leading to decreased mitochondrial respiration, membrane hyperpolarization, and augmented oxidant production compared with healthy subjects. This dysfunction correlates with platelet activation and hemolysis in vivo and can be recapitulated in vitro by exposing healthy platelets to hemoglobin or a complex V inhibitor. Further, reproduction of this dysfunction in vitro activates healthy platelets, an effect prevented by attenuation of mitochondrial hyperpolarization or by scavenging mitochondrial oxidants. These data identify bioenergetic dysfunction in SCD patients for the first time and establish mitochondrial hyperpolarization and oxidant generation as potential pathogenic mechanism in SCD as well as a modulator of healthy platelet function.


Subject(s)
Adenosine Triphosphatases/metabolism , Anemia, Sickle Cell/metabolism , Blood Platelets/metabolism , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Platelet Activation , Adult , Case-Control Studies , Female , Hemolysis , Humans , Male , Middle Aged , Mitochondrial Proton-Translocating ATPases , Oxygen Consumption , Platelet Aggregation , Reactive Oxygen Species/metabolism , Reproducibility of Results , Young Adult
8.
Am J Respir Cell Mol Biol ; 52(5): 622-33, 2015 May.
Article in English | MEDLINE | ID: mdl-25286244

ABSTRACT

Acute cellular rejection is a known risk factor for the development of obliterative bronchiolitis, which limits the long-term survival of lung transplant recipients. However, the T cell effector mechanisms in both of these processes remain incompletely understood. Using the mouse orthotopic lung transplant model, we investigated whether C57BL/6 T-bet(-/-) recipients of major histocompatibility complex (MHC)-mismatched BALB/c lung grafts develop rejection pathology and allospecific cytokine responses that differ from wild-type mice. T-bet(-/-) recipients demonstrated vigorous allograft rejection at 10 days, characterized by neutrophilic inflammation and predominantly CD8(+) T cells producing allospecific IL-17 and/or IFN-γ, in contrast to IFN-γ-dominant responses in WT mice. CD4(+) T cells produced IL-17 but not IFN-γ responses in T-bet(-/-) recipients, in contrast to WT controls. Costimulation blockade using anti-CD154 Ab significantly reduced allospecific CD8(+)IFN-γ(+) responses in both T-bet(-/-) and WT mice but had no attenuating effect on lung rejection pathology in T-bet(-/-) recipients or on the development of obliterative airway inflammation that occurred only in T-bet(-/-) recipients. However, neutralization of IL-17A significantly attenuated costimulation blockade-resistant rejection pathology and airway inflammation in T-bet(-/-) recipients. In addition, CXCL1 (neutrophil chemokine) was increased in T-bet(-/-) allografts, and IL-17 induced CXCL1 from mouse lung epithelial cells in vitro. Taken together, our data show that T-bet-deficient recipients of complete MHC-mismatched lung allografts develop costimulation blockade-resistant rejection characterized by neutrophilia and obliterative airway inflammation that is predominantly mediated by CD8(+)IL-17(+) T cells. Our data support T-bet-deficient mouse recipients of lung allografts as a viable animal model to study the immunopathogenesis of small airway injury in lung transplantation.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Graft Rejection/etiology , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Lung Transplantation/adverse effects , Lung/metabolism , Neutrophils/metabolism , Pneumonia/etiology , T-Box Domain Proteins/metabolism , Acute Disease , Allografts , Animals , Antibodies/pharmacology , CD40 Ligand/immunology , CD40 Ligand/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte , Disease Models, Animal , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Rejection/prevention & control , Histocompatibility , Inflammation Mediators/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/immunology , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/prevention & control , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics
9.
J Biol Chem ; 288(40): 28824-30, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23960074

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) is a key regulator of cell growth and metabolism. Its activity is controlled by various types of signals, including growth factors, nutrients, and stresses. In this study, we show that changes in expression levels of two antiapoptotic proteins, Bcl-2 and Bcl-XL, also affect mTORC1 signaling activity. In cells overexpressing Bcl-XL, mTORC1 activity is increased and becomes less sensitive to growth factor or nutrient conditions. In contrast, reduction in expression levels of the two antiapoptotic proteins inhibits mTORC1 signaling activity. Our results suggest that the effect of Bcl-2 and Bcl-XL on mTORC1 is mediated by FKBP38, an inhibitor of mTORC1. The two proteins compete with mTORC1 for FKBP38 binding and hence alter mTORC1 activity. This study reveals a novel cross-talk between Bcl-2/XL and mTORC1 signaling, which is likely to contribute to cancer development.


Subject(s)
Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , bcl-X Protein/metabolism , Apoptosis , Down-Regulation , Gene Knockdown Techniques , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Binding , Protein Transport , Tacrolimus Binding Proteins/metabolism
10.
Respiration ; 85(4): 267-78, 2013.
Article in English | MEDLINE | ID: mdl-23428562

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a pulmonary syndrome with growing prevalence and high mortality and morbidity that increase with age. There is no current therapy able to restore pulmonary function in ARDS patients. Preclinical models of ARDS have demonstrated that intratracheal or systemic administration of mesenchymal stem cells (MSCs) protects the lung against injury. The mechanisms responsible for the protective effects are multiple, including the secretion of multiple paracrine factors capable of modulating the immune response and restoring epithelial and endothelial integrity. Recent studies have demonstrated that MSCs can also control oxidative stress, transfer functional mitochondria to the damaged cells, and control bacterial infection by secretion of antibacterial peptides. These characteristics make MSCs promising candidates for ARDS therapy.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Respiratory Distress Syndrome/therapy , Humans , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/pathology
11.
Sci Rep ; 11(1): 490, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436736

ABSTRACT

Experimental animal models to predict physiological responses to injury and stress in humans have inherent limitations. Therefore, the development of preclinical human models is of paramount importance. Ex vivo lung perfusion (EVLP) has typically been used to recondition donor lungs before transplantation. However, this technique has recently advanced into a model to emulate lung mechanics and physiology during injury. In the present study, we propose that the EVLP of diseased human lungs is a well-suited preclinical model for translational research on chronic lung diseases. Throughout this paper, we demonstrate this technique's feasibility in pulmonary arterial hypertension (PAH), idiopathic pulmonary fibrosis (IPF), emphysema, and non-disease donor lungs not suitable for transplantation. In this study, we aimed to perfuse the lungs for 6 h with the EVLP system. This facilitated a robust and continuous assessment of airway mechanics, pulmonary hemodynamics, gas exchange, and biochemical parameters. We then collected at different time points tissue biopsies of lung parenchyma to isolate RNA and DNA to identify each disease's unique gene expression. Thus, demonstrating that EVLP could successfully serve as a clinically relevant experimental model to derive essential insights into pulmonary pathophysiology and various human lung diseases.


Subject(s)
Extracorporeal Circulation/methods , Lung Diseases/physiopathology , Lung Transplantation , Lung/physiology , Organ Preservation/standards , Tissue Donors/supply & distribution , Case-Control Studies , Female , Humans , Male , Middle Aged , Perfusion
12.
Nat Struct Mol Biol ; 12(11): 972-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16228008

ABSTRACT

The ubiquitin ligases c-Cbl and Cbl-b play a crucial role in receptor downregulation by mediating multiple monoubiquitination of receptors and promoting their sorting for lysosomal degradation. Their function is modulated through interactions with regulatory proteins including CIN85 and PIX, which recognize a proline-arginine motif in Cbl and thus promote or inhibit receptor endocytosis. We report the structures of SH3 domains of CIN85 and beta-PIX in complex with a proline-arginine peptide from Cbl-b. Both structures reveal a heterotrimeric complex containing two SH3 domains held together by a single peptide. Trimerization also occurs in solution and is facilitated by the pseudo-symmetrical peptide sequence. Moreover, ternary complexes of CIN85 and Cbl are formed in vivo and are important for the ability of Cbl to promote epidermal growth factor receptor (EGFR) downregulation. These results provide molecular explanations for a novel mechanism by which Cbl controls receptor downregulation.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Cell Cycle Proteins/chemistry , Down-Regulation , Endocytosis/physiology , Guanine Nucleotide Exchange Factors/chemistry , Models, Molecular , Proto-Oncogene Proteins c-cbl/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Calorimetry , Cell Cycle Proteins/metabolism , Crystallization , Endocytosis/genetics , ErbB Receptors/metabolism , Escherichia coli , Guanine Nucleotide Exchange Factors/metabolism , Humans , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Rho Guanine Nucleotide Exchange Factors , Structure-Activity Relationship
13.
BMJ Open Respir Res ; 6(1): e000308, 2019.
Article in English | MEDLINE | ID: mdl-30713713

ABSTRACT

Introduction: Bone marrow-derived multipotent adult progenitor cells (MAPCs) are adult allogeneic adherent stem cells currently investigated clinically for use in acute respiratory distress syndrome (ARDS). To date, there is no agreement on which is the best method for stem cells delivery in ARDS. Here, we compared the efficacy of two different methods of administration and biodistribution of MAPC for the treatment of ARDS in a sheep model. Methods: MAPC were labelled with [18F] fluoro-29-deoxy-D-glucose and delivered by endobronchial (EB) or intravenous route 1 hour after lipopolysaccharide infusion in sheep mechanically ventilated. PET/CT images were acquired to determine the biodistribution and retention of the cells at 1 and 5 hours of administration. Results: The distribution and retention of the MAPC was dependent on the method of cell administration. By EB route, PET images showed that MAPC remained at the site of administration and no changes were observed after 5 hours, whereas with intravenous route, the cells had broad biodistribution to different organs, being the lung the main organ of retention at 1 and 5 hours. MAPC demonstrated an equal effect on arterial oxygenation recovery by either route of administration. Conclusion: The EB or intravenous routes of administration of MAPC are both effective for the treatment of ARDS in an acute sheep model, and the effect of MAPC therapy is not dependent of parenchymal integration or systemic biodistribution.


Subject(s)
Adult Stem Cells/transplantation , Multipotent Stem Cells/transplantation , Respiratory Distress Syndrome/therapy , Animals , Bronchi , Cells, Cultured , Disease Models, Animal , Female , Humans , Infusions, Intravenous , Lipopolysaccharides/immunology , Male , Positron Emission Tomography Computed Tomography , Primary Cell Culture , Respiratory Distress Syndrome/diagnostic imaging , Respiratory Distress Syndrome/immunology , Sheep , Treatment Outcome
14.
Stem Cell Res Ther ; 9(1): 257, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30257725

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease for which age is the most important risk factor. Different mechanisms associated with aging, including stem cell dysfunction, have been described to participate in the pathophysiology of IPF. We observed an extrapulmonary effect associated with IPF: increase in cell senescence of bone marrow-derived mesenchymal stem cells (B-MSCs). METHODS: B-MSCs were obtained from vertebral bodies procured from IPF patients and age-matched normal controls. Cell senescence was determined by cell proliferation and expression of markers of cell senescence p16INK4A, p21, and ß-galactosidase activity. Mitochondrial function and DNA damage were measured. Paracrine induction of senescence and profibrotic responses were analyzed in vitro using human lung fibroblasts. The reparative capacity of B-MSCs was examined in vivo using the bleomycin-induced lung fibrosis model. RESULTS: In our study, we demonstrate for the first time that B-MSCs from IPF patients are senescent with significant differences in mitochondrial function, with accumulation of DNA damage resulting in defects in critical cell functions when compared with age-matched controls. Senescent IPF B-MSCs have the capability of paracrine senescence by inducing senescence in normal-aged fibroblasts, suggesting a possible link between senescent B-MSCs and the late onset of the disease. IPF B-MSCs also showed a diminished capacity to migrate and were less effective in preventing fibrotic changes observed in mice after bleomycin-induced injury, increasing illness severity and proinflammatory responses. CONCLUSIONS: We describe extrapulmonary alterations in B-MSCs from IPF patients. The consequences of having senescent B-MSCs are not completely understood, but the decrease in their ability to respond to normal activation and the risk of having a negative impact on the local niche by inducing inflammation and senescence in the neighboring cells suggests a new link between B-MSC and the onset of the disease.


Subject(s)
Aging/pathology , Cellular Senescence/genetics , Idiopathic Pulmonary Fibrosis/pathology , Mesenchymal Stem Cells/pathology , Aging/genetics , Animals , Bleomycin/toxicity , Bone Marrow Cells/pathology , Cell Proliferation/genetics , DNA Damage/genetics , Fibroblasts , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Mice
15.
JCI Insight ; 3(19)2018 10 04.
Article in English | MEDLINE | ID: mdl-30282819

ABSTRACT

The acute respiratory distress syndrome (ARDS) causes an estimated 70,000 US deaths annually. Multiple pharmacologic interventions for ARDS have been tested and failed. An unmet need is a suitable laboratory human model to predictively assess emerging therapeutics on organ function in ARDS. We previously demonstrated that the small molecule BC1215 blocks actions of a proinflammatory E3 ligase-associated protein, FBXO3, to suppress NF-κB signaling in animal models of lung injury. Ex vivo lung perfusion (EVLP) is a clinical technique that maintains lung function for possible transplant after organ donation. We used human lungs unacceptable for transplant to model endotoxemic injury with EVLP for 6 hours. LPS infusion induced inflammatory injury with impaired oxygenation of pulmonary venous circulation. BC1215 treatment after LPS rescued oxygenation and decreased inflammatory cytokines in bronchoalveolar lavage. RNA sequencing transcriptomics from biopsies taken during EVLP revealed robust inflammatory gene induction by LPS with a strong signal for NF-κB-associated transcripts. BC1215 treatment reduced the LPS induction of genes associated with inflammatory and host defense gene responses by Gene Ontology (GOterm) and pathways analysis. BC1215 also significantly antagonized LPS-mediated NF-κB activity. EVLP may provide a unique human platform for preclinical study of chemical entities such as FBXO3 inhibitors on tissue physiology.


Subject(s)
Benzylamines/pharmacology , F-Box Proteins/antagonists & inhibitors , Lung/drug effects , Perfusion/methods , Pyridines/pharmacology , Respiratory Distress Syndrome/drug therapy , Adolescent , Adult , Benzylamines/therapeutic use , Drug Evaluation, Preclinical/methods , F-Box Proteins/metabolism , Female , Humans , Lipopolysaccharides/toxicity , Lung/pathology , Male , Middle Aged , Pyridines/therapeutic use , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/pathology , Signal Transduction/drug effects
16.
ASAIO J ; 63(3): 324-332, 2017.
Article in English | MEDLINE | ID: mdl-28459743

ABSTRACT

Acute respiratory distress syndrome (ARDS) is the result of a wide variety of disorders, which can be associated with different clinical disorders or systemic diseases directly affecting the lungs. Currently, the only existing therapy is limited to supportive care. In a 6 hour pilot study, we analyzed the use of the combination of both stem cell and extracorporeal membrane oxygenation (ECMO) strategies to prevent or treat severe lung injury. A total of 11 sheep were used. Five sheep received Escherichia coli endotoxin as a control group (group 1). Three sheep that received E. coli endotoxin were treated with veno-venous ECMO support in group 2. In group 3, 3 sheep received a dose of clinical grade good manufacturing practice (GMP)-produced MultiPotent Adult Progenitor cells (MAPC) intratracheally after the end of the infusion of E. coli endotoxin, followed by ECMO support. The respiratory parameters by means of blood gas results, measurements of lung injury, inflammatory responses, and integrity of the alveolar capillary barrier after the infusion of these cells were analyzed. Our data suggest that the combination of ECMO and stem cell therapy showed better histopathologic changes with less inflammation. We believe that the combination of stem cells with the ECMO treatment may be useful in future studies investigating the diagnosis, treatment, and prevention of ARDS.


Subject(s)
Extracorporeal Membrane Oxygenation/methods , Mesenchymal Stem Cell Transplantation , Respiratory Distress Syndrome/therapy , Animals , Disease Models, Animal , Pilot Projects , Sheep
17.
PLoS One ; 10(7): e0132007, 2015.
Article in English | MEDLINE | ID: mdl-26147848

ABSTRACT

Asthma, a chronic inflammatory airway disease, is typified by high levels of TH2-cytokines and excessive generation of reactive nitrogen and oxygen species, which contribute to bronchial epithelial injury and airway remodeling. While immune function plays a major role in the pathogenesis of the disease, accumulating evidence suggests that altered cellular metabolism is a key determinant in the predisposition and disease progression of asthma. Further, several studies demonstrate altered mitochondrial function in asthmatic airways and suggest that these changes may be systemic. However, it is unknown whether systemic metabolic changes can be detected in circulating cells in asthmatic patients. Platelets are easily accessible blood cells that are known to propagate airway inflammation in asthma. Here we perform a bioenergetic screen of platelets from asthmatic and healthy individuals and demonstrate that asthmatic platelets show a decreased reliance on glycolytic processes and have increased tricarboxylic acid cycle activity. These data demonstrate a systemic alteration in asthma and are consistent with prior reports suggesting that oxidative phosphorylation is more efficient asthmatic individuals. The implications for this potential metabolic shift will be discussed in the context of increased oxidative stress and hypoxic adaptation of asthmatic patients. Further, these data suggest that platelets are potentially a good model for the monitoring of bioenergetic changes in asthma.


Subject(s)
Asthma/blood , Blood Platelets/metabolism , Glycolysis , Adult , Asthma/pathology , Blood Platelets/pathology , Cell Hypoxia , Female , Humans , Male , Mitochondria/metabolism , Oxidative Stress
18.
J Appl Physiol (1985) ; 118(1): 42-7, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25377885

ABSTRACT

Hypoxia can be damaging either because cells are directly sensitive to low oxygen pressure in their local microenvironment and/or because they are exposed to circulating factors systemically secreted in response to hypoxia. The conventional hypoxia model, breathing hypoxic air, does not allow one to distinguish between these local and systemic effects. Here we propose and validate a model for differentially applying local and systemic hypoxic challenges in an animal. We used parabiosis, two mice sharing circulation by surgical union through the skin, and tested the hypothesis that when one of the parabionts breathes room air and the other one is subjected to hypoxic air, both mice share systemic circulation but remain normoxic and hypoxic, respectively. We tested two common hypoxic paradigms in 10 parabiotic pairs: continuous hypoxia (10% O2) mimicking chronic lung diseases, and intermittent hypoxia (40 s, 21% O2; 20 s, 5% O2) simulating sleep apnea. Arterial oxygen saturation and oxygen partial pressure at muscle tissue were measured in both parabionts. Effective cross-circulation was assessed by intraperitoneally injecting a dye in one of the parabionts and measuring blood dye concentration in both animals after 2 h. The results confirmed the hypothesis that tissues of the parabiont under room air were perfused with normally oxygenated blood and, at the same time, were exposed to all of the systemic mediators secreted by the other parabiont actually subjected to hypoxia. In conclusion, combination of parabiosis and hypoxic/normoxic air breathing is a novel approach to investigate the effects of local and systemic hypoxia in respiratory diseases.


Subject(s)
Hypoxia/physiopathology , Oxygen/blood , Parabiosis , Animals , Disease Models, Animal , Hypoxia/blood , Mice , Respiration
19.
J Mech Behav Biomed Mater ; 37: 186-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24946269

ABSTRACT

Lung disease models are useful to study how cell engraftment, proliferation and differentiation are modulated in lung bioengineering. The aim of this work was to characterize the local stiffness of decellularized lungs in aged and fibrotic mice. Mice (2- and 24-month old; 14 of each) with lung fibrosis (N=20) and healthy controls (N=8) were euthanized after 11 days of intratracheal bleomycin (fibrosis) or saline (controls) infusion. The lungs were excised, decellularized by a conventional detergent-based (sodium-dodecyl sulfate) procedure and slices of the acellular lungs were prepared to measure the local stiffness by means of atomic force microscopy. The local stiffness of the different sites in acellular fibrotic lungs was very inhomogeneous within the lung and increased according to the degree of the structural fibrotic lesion. Local stiffness of the acellular lungs did not show statistically significant differences caused by age. The group of mice most affected by fibrosis exhibited local stiffness that were ~2-fold higher than in the control mice: from 27.2±1.64 to 64.8±7.1kPa in the alveolar septa, from 56.6±4.6 to 99.9±11.7kPa in the visceral pleura, from 41.1±8.0 to 105.2±13.6kPa in the tunica adventitia, and from 79.3±7.2 to 146.6±28.8kPa in the tunica intima. Since acellular lungs from mice with bleomycin-induced fibrosis present considerable micromechanical inhomogeneity, this model can be a useful tool to better investigate how different degrees of extracellular matrix lesion modulate cell fate in the process of organ bioengineering from decellularized lungs.


Subject(s)
Extracellular Matrix/metabolism , Lung/pathology , Mechanical Phenomena , Tissue Scaffolds , Aging/pathology , Animals , Biomechanical Phenomena , Bleomycin/adverse effects , Collagen/metabolism , Extracellular Matrix/drug effects , Female , Fibrosis , Lung/drug effects , Mice , Mice, Inbred C57BL
20.
Stem Cell Res Ther ; 5(2): 42, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24670268

ABSTRACT

INTRODUCTION: Acute respiratory distress syndrome (ARDS) is the most common cause of respiratory failure among critically ill subjects, sepsis and severe bacterial pneumonia being its most common causes. The only interventions that have proven beneficial are protective ventilation strategies and fluid conservation approaches. New therapies are needed to address this common clinical problem. Others and we have previously shown the beneficial effect of infusion of exogenous adult stem cells in different pre-clinical models of ARDS. METHODS: In the present study endotoxin was infused intravenously into 14 sheep from which 6 received different doses of adult stem cells by intrabronchial delivery to evaluate the effect of stem cell therapy. RESULTS: After administration of endotoxin, there was a rapid decline in oxygenation to hypoxemic values, indicative of severe-to-moderate ARDS. None of the animals treated with saline solution recovered to normal baseline values during the 6 hours that the animals were followed. In contrast, sheep treated with a dose of 40 million adult stem cells returned their levels of oxygen in their blood to baseline two hours after the cells were infused. Similarly, improvements in carbon dioxide (CO2) clearance, pulmonary vascular pressures and inflammation were observed and confirmed by histology and by the decrease in lung edema. CONCLUSIONS: We concluded that instillation of adult non-hematopoietic stem cells can diminish the impact of endotoxin and accelerate recovery of oxygenation, CO2 removal and inflammation in the ovine model, making the use of adult stem cells a real alternative for future therapies for ARDS.


Subject(s)
Adult Stem Cells/cytology , Bone Marrow Cells/cytology , Bone Marrow Transplantation/methods , Respiratory Distress Syndrome/therapy , Stem Cell Transplantation/methods , Animals , Disease Models, Animal , Humans , Lipopolysaccharides , Respiratory Distress Syndrome/chemically induced , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL