Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lea Barthel is active.

Publication


Featured researches published by Lea Barthel.


Nature | 2014

Muc5b is required for airway defence

Michelle G. Roy; Alessandra Livraghi-Butrico; Ashley A. Fletcher; Melissa M. McElwee; Scott E. Evans; Ryan M. Boerner; Samantha N. Alexander; Lindsey K. Bellinghausen; Alfred S. Song; Youlia Petrova; Michael J. Tuvim; Roberto Adachi; Irlanda Romo; Andrea S. Bordt; M. Gabriela Bowden; Joseph H. Sisson; Prescott G. Woodruff; David J. Thornton; Karine Rousseau; Maria Miguelina De La Garza; Seyed Javad Moghaddam; Harry Karmouty-Quintana; Michael R. Blackburn; Scott M. Drouin; C. William Davis; Kristy A. Terrell; Barbara R. Grubb; Wanda K. O'Neal; Sonia C. Flores; Adela Cota-Gomez

Respiratory surfaces are exposed to billions of particulates and pathogens daily. A protective mucus barrier traps and eliminates them through mucociliary clearance (MCC). However, excessive mucus contributes to transient respiratory infections and to the pathogenesis of numerous respiratory diseases. MUC5AC and MUC5B are evolutionarily conserved genes that encode structurally related mucin glycoproteins, the principal macromolecules in airway mucus. Genetic variants are linked to diverse lung diseases, but specific roles for MUC5AC and MUC5B in MCC, and the lasting effects of their inhibition, are unknown. Here we show that mouse Muc5b (but not Muc5ac) is required for MCC, for controlling infections in the airways and middle ear, and for maintaining immune homeostasis in mouse lungs, whereas Muc5ac is dispensable. Muc5b deficiency caused materials to accumulate in upper and lower airways. This defect led to chronic infection by multiple bacterial species, including Staphylococcus aureus, and to inflammation that failed to resolve normally. Apoptotic macrophages accumulated, phagocytosis was impaired, and interleukin-23 (IL-23) production was reduced in Muc5b−/− mice. By contrast, in mice that transgenically overexpress Muc5b, macrophage functions improved. Existing dogma defines mucous phenotypes in asthma and chronic obstructive pulmonary disease (COPD) as driven by increased MUC5AC, with MUC5B levels either unaffected or increased in expectorated sputum. However, in many patients, MUC5B production at airway surfaces decreases by as much as 90%. By distinguishing a specific role for Muc5b in MCC, and by determining its impact on bacterial infections and inflammation in mice, our results provide a refined framework for designing targeted therapies to control mucin secretion and restore MCC.


Nature Medicine | 2012

The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis

Eric P. Schmidt; Yimu Yang; William J. Janssen; Aneta Gandjeva; Mario J. Perez; Lea Barthel; Rachel L. Zemans; Joel Bowman; Dan Koyanagi; Zulma X. Yunt; Lynelle P. Smith; Sara S Cheng; Katherine H. Overdier; Kathy Thompson; Mark W. Geraci; Ivor S. Douglas; David B. Pearse; Rubin M. Tuder

Sepsis, a systemic inflammatory response to infection, commonly progresses to acute lung injury (ALI), an inflammatory lung disease with high morbidity. We postulated that sepsis-associated ALI is initiated by degradation of the pulmonary endothelial glycocalyx, leading to neutrophil adherence and inflammation. Using intravital microscopy, we found that endotoxemia in mice rapidly induced pulmonary microvascular glycocalyx degradation via tumor necrosis factor-α (TNF-α)-dependent mechanisms. Glycocalyx degradation involved the specific loss of heparan sulfate and coincided with activation of endothelial heparanase, a TNF-α–responsive, heparan sulfate–specific glucuronidase. Glycocalyx degradation increased the availability of endothelial surface adhesion molecules to circulating microspheres and contributed to neutrophil adhesion. Heparanase inhibition prevented endotoxemia-associated glycocalyx loss and neutrophil adhesion and, accordingly, attenuated sepsis-induced ALI and mortality in mice. These findings are potentially relevant to human disease, as sepsis-associated respiratory failure in humans was associated with higher plasma heparan sulfate degradation activity; moreover, heparanase content was higher in human lung biopsies showing diffuse alveolar damage than in normal human lung tissue.


American Journal of Respiratory and Critical Care Medicine | 2011

Fas Determines Differential Fates of Resident and Recruited Macrophages during Resolution of Acute Lung Injury

William J. Janssen; Lea Barthel; Alaina Muldrow; Rebecca E. Oberley-Deegan; Mark T. Kearns; Claudia V. Jakubzick; Peter M. Henson

RATIONALE During acute lung injury (ALI) the macrophage pool expands markedly as inflammatory monocytes migrate from the circulation to the airspaces. As inflammation resolves, macrophage numbers return to preinjury levels and normal tissue structure and function are restored. OBJECTIVES To determine the fate of resident and recruited macrophages during the resolution of ALI in mice and to elucidate the mechanisms responsible for macrophage removal. METHODS ALI was induced in mice using influenza A (H1N1; PR8) infection and LPS instillation. Dye labeling techniques, bone marrow transplantation, and surface immunophenotyping were used to distinguish resident and recruited macrophages during inflammation and to study the role of Fas in determining macrophage fate during resolving ALI. MEASUREMENTS AND MAIN RESULTS During acute and resolving lung injury from influenza A and LPS, a high proportion of the original resident alveolar macrophages persisted. In contrast, recruited macrophages exhibited robust accumulation in early inflammation, followed by a progressive decline in their number. This decline was mediated by apoptosis with local phagocytic clearance. Recruited macrophages expressed high levels of the death receptor Fas and were rapidly depleted from the airspaces by Fas-activating antibodies. In contrast, macrophage depletion was inhibited in mice treated with Fas-blocking antibodies and in chimeras with Fas-deficient bone marrow. Caspase-8 inhibition prevented macrophage apoptosis and delayed the resolution of ALI. CONCLUSIONS These findings indicate that Fas-induced apoptosis of recruited macrophages is essential for complete resolution of ALI.


Science Translational Medicine | 2009

Tetracyclines That Promote SMN2 Exon 7 Splicing as Therapeutics for Spinal Muscular Atrophy

Michelle L. Hastings; Joel Berniac; Ying Hsiu Liu; Paul Abato; Francine M. Jodelka; Lea Barthel; Sujatha Kumar; Caroline Dudley; Mark L. Nelson; Kelley Larson; Jason Edmonds; Todd E. Bowser; Michael Draper; Paul Higgins; Adrian R. Krainer

Tetracycline derivatives increase exon 7 splicing during RNA processing of the spinal muscular atrophy–modifying gene SMN2, which may prove therapeutically useful. Correcting Splicing to Prevent Spinal Muscular Atrophy Even before birth, a child with the most severe form of spinal muscular atrophy (SMA) may be frail, so that the mother feels only faint fetal movements during the last months of pregnancy. After birth, children with this autosomal recessive disease exhibit weak muscles, swallowing difficulties, and respiratory problems, often dying within the first 2 years of life. These symptoms are caused by the degeneration of certain motor neurons in the spinal cord, resulting in muscle weakness and shrinking. There is no cure or effective therapy for SMA, which kills more infants than any other genetic disorder. Now, Krainer and colleagues describe a compound with promise as an SMA therapeutic. SMA is usually caused by loss-of-function mutations in the SMN1 gene, which encodes a protein that aids in the assembly of the spliceosome. This large protein-RNA complex removes introns from RNA transcripts to create mature mRNAs. An inadequate amount of SMN protein causes SMA, but it is not clear why the deficiency selectively affects spinal cord motor neurons. Cells contain a second source of the SMN protein, the SMN2 gene. Because of a single nucleotide change in SMN2, its RNA transcript is usually spliced incorrectly, such that exon 7 is left out; the encoded truncated protein is quite unstable. Thus, one way to treat SMA would be to correct SMN2 splicing. Although compounds have been identified that increase the production of full-length SMN protein from SMN2, they act in a relatively nonspecific manner; some are toxic. A better possibility might be a drug that specifically alters splicing, potentially reducing side effects. The Hastings and Krainer labs, in collaboration with Paratek Pharmaceuticals, sought to identify a small molecule that specifically improves exon 7 splicing of SMN2 RNA in a cell-free assay. In a screen, they found one compound, a tetracycline derivative called PTK-SMA1, that could do this. PTK-SMA1 appears to stimulate exon 7 splicing in SMN1/2 specifically, not affecting splicing of other tested substrates. The researchers determined that PTK-SMA1 increases SMN protein concentrations both in fibroblasts derived from an SMA patient and in mouse models of SMA. Because PTK-SMA1 does not cross the blood-brain barrier but would need to do so in order to be therapeutically useful, the researchers now aim to modify PTK-SMA1 to achieve that end. Furthermore, as RNA splicing defects may contribute to other diseases, additional tetracycline derivatives that can repair specific splicing defects could potentially be identified and prove useful. There is at present no cure or effective therapy for spinal muscular atrophy (SMA), a neurodegenerative disease that is the leading genetic cause of infant mortality. SMA usually results from loss of the SMN1 (survival of motor neuron 1) gene, which leads to selective motor neuron degeneration. SMN2 is nearly identical to SMN1 but has a nucleotide replacement that causes exon 7 skipping, resulting in a truncated, unstable version of the SMA protein. SMN2 is present in all SMA patients, and correcting SMN2 splicing is a promising approach for SMA therapy. We identified a tetracycline-like compound, PTK-SMA1, which stimulates exon 7 splicing and increases SMN protein levels in vitro and in vivo in mice. Unlike previously identified molecules that stimulate SMN production via SMN2 promoter activation or undefined mechanisms, PTK-SMA1 is a unique therapeutic candidate in that it acts by directly stimulating splicing of exon 7. Synthetic small-molecule compounds such as PTK-SMA1 offer an alternative to antisense oligonucleotide therapies that are being developed as therapeutics for a number of disease-associated splicing defects.


American Journal of Physiology-renal Physiology | 2012

Macrophages mediate lung inflammation in a mouse model of ischemic acute kidney injury.

Christopher Altmann; Ana Andres-Hernando; Rachel H. McMahan; Nilesh Ahuja; Zhibin He; Chris J. Rivard; Charles L. Edelstein; Lea Barthel; William J. Janssen; Sarah Faubel

Serum IL-6 is increased in acute kidney injury (AKI) and inhibition of IL-6 reduces AKI-mediated lung inflammation. We hypothesized that circulating monocytes produce IL-6 and that alveolar macrophages mediate lung inflammation after AKI via chemokine (CXCL1) production. To investigate systemic and alveolar macrophages in lung injury after AKI, sham operation or 22 min of renal pedicle clamping (AKI) was performed in three experimental settings: 1) systemic macrophage depletion via diphtheria toxin (DT) injection to CD11b-DTR transgenic mice, 2) DT injection to wild-type mice, and 3) alveolar macrophage depletion via intratracheal (IT) liposome-encapsulated clodronate (LEC) administration to wild-type mice. In mice with AKI and systemic macrophage depletion (CD11b-DTR transgenic administered DT) vs. vehicle-treated AKI, blood monocytes and lung interstitial macrophages were reduced, renal function was similar, serum IL-6 was increased, lung inflammation was improved, lung CXCL1 was reduced, and lung capillary leak was increased. In wild-type mice with AKI administered DT vs. vehicle, serum IL-6 was increased. In mice with AKI and alveolar macrophage depletion (IT-LEC) vs. AKI with normal alveolar macrophage content, blood monocytes and lung interstitial macrophages were similar, alveolar macrophages were reduced, renal function was similar, lung inflammation was improved, lung CXCL1 was reduced, and lung capillary leak was increased. In conclusion, administration of DT in AKI is proinflammatory, limiting the use of the DTR-transgenic model to study systemic effects of AKI. Mice with AKI and either systemic mononuclear phagocyte depletion or alveolar macrophage depletion had reduced lung inflammation and lung CXCL1, but increased lung capillary leak; thus, mononuclear phagocytes mediate lung inflammation, but they protect against lung capillary leak after ischemic AKI. Since macrophage activation and chemokine production are key events in the development of acute lung injury (ALI), these data provide further evidence that AKI may cause ALI.


American Journal of Respiratory and Critical Care Medicine | 2015

The Causal Role of IL-4 and IL-13 in Schistosoma mansoni Pulmonary Hypertension

Rahul Kumar; Claudia Mickael; Jacob J. Chabon; Liya Gebreab; Alleluiah Rutebemberwa; Alexandra Rodriguez Garcia; Daniel E. Koyanagi; Linda Sanders; Aneta Gandjeva; Mark T. Kearns; Lea Barthel; William J. Janssen; Thais Mauad; Angela Bandeira; Eric P. Schmidt; Rubin M. Tuder; Brian B. Graham

RATIONALE The etiology of schistosomiasis-associated pulmonary arterial hypertension (PAH), a major cause of PAH worldwide, is poorly understood. Schistosoma mansoni exposure results in prototypical type-2 inflammation. Furthermore, transforming growth factor (TGF)-β signaling is required for experimental pulmonary hypertension (PH) caused by Schistosoma exposure. OBJECTIVES We hypothesized type-2 inflammation driven by IL-4 and IL-13 is necessary for Schistosoma-induced TGF-β-dependent vascular remodeling. METHODS Wild-type, IL-4(-/-), IL-13(-/-), and IL-4(-/-)IL-13(-/-) mice (C57BL6/J background) were intraperitoneally sensitized and intravenously challenged with S. mansoni eggs to induce experimental PH. Right ventricular catheterization was then performed, followed by quantitative analysis of the lung tissue. Lung tissue from patients with schistosomiasis-associated and connective tissue disease-associated PAH was also systematically analyzed. MEASUREMENTS AND MAIN RESULTS Mice with experimental Schistosoma-induced PH had evidence of increased IL-4 and IL-13 signaling. IL-4(-/-)IL-13(-/-) mice, but not single knockout IL-4(-/-) or IL-13(-/-) mice, were protected from Schistosoma-induced PH, with decreased right ventricular pressures, pulmonary vascular remodeling, and right ventricular hypertrophy. IL-4(-/-)IL-13(-/-) mice had less pulmonary vascular phospho-signal transducer and activator of transcription 6 (STAT6) and phospho-Smad2/3 activity, potentially caused by decreased TGF-β activation by macrophages. In vivo treatment with a STAT6 inhibitor and IL-4(-/-)IL-13(-/-) bone marrow transplantation also protected against Schistosoma-PH. Lung tissue from patients with schistosomiasis-associated and connective tissue disease-associated PAH had evidence of type-2 inflammation. CONCLUSIONS Combined IL-4 and IL-13 deficiency is required for protection against TGF-β-induced pulmonary vascular disease after Schistosoma exposure, and targeted inhibition of this pathway is a potential novel therapeutic approach for patients with schistosomiasis-associated PAH.


American Journal of Respiratory Cell and Molecular Biology | 2017

Cell Origin Dictates Programming of Resident versus Recruited Macrophages during Acute Lung Injury

Kara J. Mould; Lea Barthel; Michael P. Mohning; Stacey M. Thomas; Alexandra L. McCubbrey; Thomas Danhorn; Sonia M. Leach; Tasha E. Fingerlin; Brian P. O’Connor; Julie A. Reisz; Angelo D’Alessandro; Donna L. Bratton; Claudia V. Jakubzick; William J. Janssen

&NA; Two populations of alveolar macrophages (AMs) coexist in the inflamed lung: resident AMs that arise during embryogenesis, and recruited AMs that originate postnatally from circulating monocytes. The objective of this study was to determine whether origin or environment dictates the transcriptional, metabolic, and functional programming of these two ontologically distinct populations over the time course of acute inflammation. RNA sequencing demonstrated marked transcriptional differences between resident and recruited AMs affecting three main areas: proliferation, inflammatory signaling, and metabolism. Functional assays and metabolomic studies confirmed these differences and demonstrated that resident AMs proliferate locally and are governed by increased tricarboxylic acid cycle and amino acid metabolism. Conversely, recruited AMs produce inflammatory cytokines in association with increased glycolytic and arginine metabolism. Collectively, the data show that even though they coexist in the same environment, inflammatory macrophage subsets have distinct immunometabolic programs and perform specialized functions during inflammation that are associated with their cellular origin.


Nature Communications | 2017

TGF-β activation by bone marrow-derived thrombospondin-1 causes Schistosoma - and hypoxia-induced pulmonary hypertension

Rahul Kumar; Claudia Mickael; Biruk Kassa; Liya Gebreab; Jeffrey C. Robinson; Daniel E. Koyanagi; Linda Sanders; Lea Barthel; Christina A. Meadows; Daniel Fox; David M. Irwin; Min Li; B. Alexandre McKeon; Suzette R. Riddle; R. Dale Brown; Leslie E. Morgan; Christopher M. Evans; Daniel Hernandez-Saavedra; Angela Bandeira; James P. Maloney; Todd M. Bull; William J. Janssen; Kurt R. Stenmark; Rubin M. Tuder; Brian B. Graham

Pulmonary arterial hypertension (PAH) is an obstructive disease of the precapillary pulmonary arteries. Schistosomiasis-associated PAH shares altered vascular TGF-β signalling with idiopathic, heritable and autoimmune-associated etiologies; moreover, TGF-β blockade can prevent experimental pulmonary hypertension (PH) in pre-clinical models. TGF-β is regulated at the level of activation, but how TGF-β is activated in this disease is unknown. Here we show TGF-β activation by thrombospondin-1 (TSP-1) is both required and sufficient for the development of PH in Schistosoma-exposed mice. Following Schistosoma exposure, TSP-1 levels in the lung increase, via recruitment of circulating monocytes, while TSP-1 inhibition or knockout bone marrow prevents TGF-β activation and protects against PH development. TSP-1 blockade also prevents the PH in a second model, chronic hypoxia. Lastly, the plasma concentration of TSP-1 is significantly increased in subjects with scleroderma following PAH development. Targeting TSP-1-dependent activation of TGF-β could thus be a therapeutic approach in TGF-β-dependent vascular diseases.


Journal of Immunological Methods | 2010

Development and characterization of a lung-protective method of bone marrow transplantation in the mouse

William J. Janssen; Alaina Muldrow; Mark T. Kearns; Lea Barthel; Peter M. Henson

Allogeneic bone marrow transplantation is a common method used to study the contribution of myeloid and lymphoid cell populations in murine models of disease. The method requires lethal doses of radiation to ablate the bone marrow. Unintended consequences of radiation include organ injury and inflammatory cell activation. The goal of our study was to determine the degree to which bone marrow transplantation alters lungs and to develop a system to protect the lungs during radiation. C57BL/6 mice were subjected to total body irradiation with 900cGy and then transplanted with bone marrow from green fluorescent protein (GFP) expressing mice. Resultant chimeras exhibited a significant decline in alveolar macrophage numbers within 72h, modest influx of neutrophils in the lungs at 14days, and repopulation of the lungs by alveolar macrophages of bone marrow origin by 28days. Neutrophil influx and alveolar macrophage turnover were prevented when 1cm thick lead shields were used to protect the lungs during radiation, such that 8weeks after transplantation less than 30% of alveolar macrophages were of donor origin. Lung-shielded mice achieved a high level of bone marrow engraftment with greater than 95% of circulating leukocytes expressing GFP. In addition, their response to intratracheal lipopolysaccharide was similar to non-transplanted mice. We describe a model whereby lead shields protect resident cell populations in the lungs from radiation during bone marrow transplantation but permit full bone marrow engraftment. This system may be applicable to other organ systems in which protection from radiation during bone marrow transplantation is desired.


Science Translational Medicine | 2017

Neutrophil transfer of miR-223 to lung epithelial cells dampens acute lung injury in mice

Viola Neudecker; Eric T. Clambey; Eric P. Schmidt; Thomas A. Packard; Bennett Davenport; Theodore J. Standiford; Tingting Weng; Ashley A. Fletcher; Lea Barthel; Joanne C. Masterson; Glenn T. Furuta; Chunyan Cai; Michael R. Blackburn; Adit A. Ginde; Michael W. Graner; William J. Janssen; Rachel L. Zemans; Christopher M. Evans; Ellen L. Burnham; Dirk Homann; Marc Moss; Simone Kreth; Kai Zacharowski; Peter M. Henson; Holger K. Eltzschig

Intercellular transfer of miR-223 from neutrophils to alveolar epithelial cells reduces lung inflammation in a mouse model of ventilator-induced lung injury or pulmonary bacterial infection. Starting an intercellular conversation In a new study, Neudecker et al. show that transfer of microRNA-223 (miR-223) from neutrophils to lung alveoli helps to dampen lung inflammation and promotes the resolution of ventilator-induced lung injury in mice. The authors suggest that neutrophils secrete microRNAs in microvesicles that are then taken up by alveolar epithelial cells. They show that miR-223–deficient mice are prone to lung injury, whereas overexpression of miR-223 is protective. Intercellular transfer of microRNAs can mediate communication between critical effector cells. We hypothesized that transfer of neutrophil-derived microRNAs to pulmonary epithelial cells could alter mucosal gene expression during acute lung injury. Pulmonary-epithelial microRNA profiling during coculture of alveolar epithelial cells with polymorphonuclear neutrophils (PMNs) revealed a selective increase in lung epithelial cell expression of microRNA-223 (miR-223). Analysis of PMN-derived supernatants showed activation-dependent release of miR-223 and subsequent transfer to alveolar epithelial cells during coculture in vitro or after ventilator-induced acute lung injury in mice. Genetic studies indicated that miR-223 deficiency was associated with severe lung inflammation, whereas pulmonary overexpression of miR-223 in mice resulted in protection during acute lung injury induced by mechanical ventilation or by infection with Staphylococcus aureus. Studies of putative miR-223 gene targets implicated repression of poly(adenosine diphosphate–ribose) polymerase–1 (PARP-1) in the miR-223–dependent attenuation of lung inflammation. Together, these findings suggest that intercellular transfer of miR-223 from neutrophils to pulmonary epithelial cells may dampen acute lung injury through repression of PARP-1.

Collaboration


Dive into the Lea Barthel's collaboration.

Top Co-Authors

Avatar

William J. Janssen

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Peter M. Henson

Indiana University Bloomington

View shared research outputs
Top Co-Authors

Avatar

Mark T. Kearns

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Michael P. Mohning

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Rubin M. Tuder

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Zulma X. Yunt

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Eric P. Schmidt

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Kara J. Mould

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Alaina Muldrow

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge