Network


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

Hotspot


Dive into the research topics where Lynn C. Welch is active.

Publication


Featured researches published by Lynn C. Welch.


Journal of Clinical Investigation | 2008

AMP-activated protein kinase regulates CO2-induced alveolar epithelial dysfunction in rats and human cells by promoting Na,K-ATPase endocytosis

István Vadász; Laura A. Dada; Arturo Briva; Humberto E. Trejo; Lynn C. Welch; Jiwang Chen; Peter T. Toth; Emilia Lecuona; Lee A. Witters; Paul T. Schumacker; Navdeep S. Chandel; Werner Seeger; Jacob I. Sznajder

Hypercapnia (elevated CO(2) levels) occurs as a consequence of poor alveolar ventilation and impairs alveolar fluid reabsorption (AFR) by promoting Na,K-ATPase endocytosis. We studied the mechanisms regulating CO(2)-induced Na,K-ATPase endocytosis in alveolar epithelial cells (AECs) and alveolar epithelial dysfunction in rats. Elevated CO(2) levels caused a rapid activation of AMP-activated protein kinase (AMPK) in AECs, a key regulator of metabolic homeostasis. Activation of AMPK was mediated by a CO(2)-triggered increase in intracellular Ca(2+) concentration and Ca(2+)/calmodulin-dependent kinase kinase-beta (CaMKK-beta). Chelating intracellular Ca(2+) or abrogating CaMKK-beta function by gene silencing or chemical inhibition prevented the CO(2)-induced AMPK activation in AECs. Activation of AMPK or overexpression of constitutively active AMPK was sufficient to activate PKC-zeta and promote Na,K-ATPase endocytosis. Inhibition or downregulation of AMPK via adenoviral delivery of dominant-negative AMPK-alpha(1) prevented CO(2)-induced Na,K-ATPase endocytosis. The hypercapnia effects were independent of intracellular ROS. Exposure of rats to hypercapnia for up to 7 days caused a sustained decrease in AFR. Pretreatment with a beta-adrenergic agonist, isoproterenol, or a cAMP analog ameliorated the hypercapnia-induced impairment of AFR. Accordingly, we provide evidence that elevated CO(2) levels are sensed by AECs and that AMPK mediates CO(2)-induced Na,K-ATPase endocytosis and alveolar epithelial dysfunction, which can be prevented with beta-adrenergic agonists and cAMP.


Circulation Research | 2004

Upregulation of Alveolar Epithelial Active Na+ Transport Is Dependent on β2-Adrenergic Receptor Signaling

Gökhan M. Mutlu; Vidas Dumasius; James Burhop; Pamela J. McShane; Fan Jing Meng; Lynn C. Welch; Andrew Dumasius; Nima Mohebahmadi; Gloria Thakuria; Karen Hardiman; Sadis Matalon; Steven M. Hollenberg; Phillip Factor

Abstract— Alveolar epithelial β-adrenergic receptor (βAR) activation accelerates active Na+ transport in lung epithelial cells in vitro and speeds alveolar edema resolution in human lung tissue and normal and injured animal lungs. Whether these receptors are essential for alveolar fluid clearance (AFC) or if other mechanisms are sufficient to regulate active transport is unknown. In this study, we report that mice with no β1- or β2-adrenergic receptors (β1AR−/−/β2AR−/−) have reduced distal lung Na,K-ATPase function and diminished basal and amiloride-sensitive AFC. Total lung water content in these animals was not different from wild-type controls, suggesting that βAR signaling may not be required for alveolar fluid homeostasis in uninjured lungs. Comparison of isoproterenol-sensitive AFC in mice with β1- but not β2-adrenergic receptors to β1AR−/−/β2AR−/− mice indicates that the β2AR mediates the bulk of β-adrenergic–sensitive alveolar active Na+ transport. To test the necessity of βAR signaling in acute lung injury, β1AR−/−/β2AR−/−, β1AR+/+/β2AR−/−, and β1AR+/+/β2AR+/+ mice were exposed to 100% oxygen for up to 204 hours. β1AR−/−/β2AR−/− and β1AR+/+/β2AR−/− mice had more lung water and worse survival from this form of acute lung injury than wild-type controls. Adenoviral-mediated rescue of β2-adrenergic receptor (β2AR) function into the alveolar epithelium of β1AR−/−/β2AR−/− and β1AR+/+/β2AR−/− mice normalized distal lung β2AR function, alveolar epithelial active Na+ transport, and survival from hyperoxia. These findings indicate that βAR signaling may not be necessary for basal AFC, and that β2AR is essential for the adaptive physiological response needed to clear excess fluid from the alveolar airspace of normal and injured lungs.


PLOS ONE | 2007

High CO2 levels impair alveolar epithelial function independently of pH.

Arturo Briva; István Vadász; Emilia Lecuona; Lynn C. Welch; Jiwang Chen; Laura A. Dada; Humberto E. Trejo; Vidas Dumasius; Zaher S. Azzam; Pavlos Myrianthefs; Daniel Batlle; Yosef Gruenbaum; Jacob I. Sznajder

Background In patients with acute respiratory failure, gas exchange is impaired due to the accumulation of fluid in the lung airspaces. This life-threatening syndrome is treated with mechanical ventilation, which is adjusted to maintain gas exchange, but can be associated with the accumulation of carbon dioxide in the lung. Carbon dioxide (CO2) is a by-product of cellular energy utilization and its elimination is affected via alveolar epithelial cells. Signaling pathways sensitive to changes in CO2 levels were described in plants and neuronal mammalian cells. However, it has not been fully elucidated whether non-neuronal cells sense and respond to CO2. The Na,K-ATPase consumes ∼40% of the cellular metabolism to maintain cell homeostasis. Our study examines the effects of increased pCO2 on the epithelial Na,K-ATPase a major contributor to alveolar fluid reabsorption which is a marker of alveolar epithelial function. Principal Findings We found that short-term increases in pCO2 impaired alveolar fluid reabsorption in rats. Also, we provide evidence that non-excitable, alveolar epithelial cells sense and respond to high levels of CO2, independently of extracellular and intracellular pH, by inhibiting Na,K-ATPase function, via activation of PKCζ which phosphorylates the Na,K-ATPase, causing it to endocytose from the plasma membrane into intracellular pools. Conclusions Our data suggest that alveolar epithelial cells, through which CO2 is eliminated in mammals, are highly sensitive to hypercapnia. Elevated CO2 levels impair alveolar epithelial function, independently of pH, which is relevant in patients with lung diseases and altered alveolar gas exchange.


Molecular and Cellular Biology | 2011

Hypoxia Leads to Na,K-ATPase Downregulation via Ca2+ Release-Activated Ca2+ Channels and AMPK Activation

Galina A. Gusarova; Humberto E. Trejo; Laura A. Dada; Arturo Briva; Lynn C. Welch; Robert B. Hamanaka; Gökhan M. Mutlu; Navdeep S. Chandel; Murali Prakriya; Jacob I. Sznajder

ABSTRACT To maintain cellular ATP levels, hypoxia leads to Na,K-ATPase inhibition in a process dependent on reactive oxygen species (ROS) and the activation of AMP-activated kinase α1 (AMPK-α1). We report here that during hypoxia AMPK activation does not require the liver kinase B1 (LKB1) but requires the release of Ca2+ from the endoplasmic reticulum (ER) and redistribution of STIM1 to ER-plasma membrane junctions, leading to calcium entry via Ca2+ release-activated Ca2+ (CRAC) channels. This increase in intracellular Ca2+ induces Ca2+/calmodulin-dependent kinase kinase β (CaMKKβ)-mediated AMPK activation and Na,K-ATPase downregulation. Also, in cells unable to generate mitochondrial ROS, hypoxia failed to increase intracellular Ca2+ concentration while a STIM1 mutant rescued the AMPK activation, suggesting that ROS act upstream of Ca2+ signaling. Furthermore, inhibition of CRAC channel function in rat lungs prevented the impairment of alveolar fluid reabsorption caused by hypoxia. These data suggest that during hypoxia, calcium entry via CRAC channels leads to AMPK activation, Na,K-ATPase downregulation, and alveolar epithelial dysfunction.


Circulation Research | 2005

Interdependency of β-Adrenergic Receptors and CFTR in Regulation of Alveolar Active Na+ Transport

Gökhan M. Mutlu; Yochai Adir; Mohammed Jameel; Alexander T. Akhmedov; Lynn C. Welch; Vidas Dumasius; Fan Jing Meng; Joseph Zabner; Craig Koenig; Erin Lewis; Rajesh Balagani; Geri L. Traver; Jacob I. Sznajder; Phillip Factor

&bgr;-Adrenergic receptors (&bgr;AR) regulate active Na+ transport in the alveolar epithelium and accelerate clearance of excess airspace fluid. Accumulating data indicates that the cystic fibrosis transmembrane conductance regulator (CFTR) is important for upregulation of the active ion transport that is needed to maintain alveolar fluid homeostasis during pulmonary edema. We hypothesized that &bgr;AR regulation of alveolar active transport may be mediated via a CFTR dependent pathway. To test this hypothesis we used a recombinant adenovirus that expresses a human CFTR cDNA (adCFTR) to increase CFTR function in the alveolar epithelium of normal rats and mice. Alveolar fluid clearance (AFC), an index of alveolar active Na+ transport, was 92% greater in CFTR overexpressing lungs than controls. Addition of the Cl− channel blockers NPPB, glibenclamide, or bumetanide and experiments using Cl− free alveolar instillate solutions indicate that the accelerated AFC in this model is due to increased Cl− channel function. Conversely, CFTR overexpression in mice with no &bgr;1- or &bgr;2-adrenergic receptors had no effect on AFC. Overexpression of a human &bgr;2AR in the alveolar epithelium significantly increased AFC in normal mice but had no effect in mice with a non-functional human CFTR gene (&Dgr;&phgr;508 mutation). These studies indicate that upregulation of alveolar CFTR function speeds clearance of excess fluid from the airspace and that CFTRs effect on active Na+ transport requires the &bgr;AR. These studies reveal a previously undetected interdependency between CFTR and &bgr;AR that is essential for upregulation of active Na+ transport and fluid clearance in the alveolus.


American Journal of Respiratory Cell and Molecular Biology | 2013

Hypercapnia impairs lung neutrophil function and increases mortality in murine Pseudomonas pneumonia

Khalilah L. Gates; Heather A. Howell; Aisha Nair; Christine U. Vohwinkel; Lynn C. Welch; Greg J. Beitel; Alan R. Hauser; Jacob I. Sznajder; Peter H. S. Sporn

Hypercapnia, an elevation of the level of carbon dioxide (CO2) in blood and tissues, is a marker of poor prognosis in chronic obstructive pulmonary disease and other pulmonary disorders. We previously reported that hypercapnia inhibits the expression of TNF and IL-6 and phagocytosis in macrophages in vitro. In the present study, we determined the effects of normoxic hypercapnia (10% CO2, 21% O2, and 69% N2) on outcomes of Pseudomonas aeruginosa pneumonia in BALB/c mice and on pulmonary neutrophil function. We found that the mortality of P. aeruginosa pneumonia was increased in 10% CO2-exposed compared with air-exposed mice. Hypercapnia increased pneumonia mortality similarly in mice with acute and chronic respiratory acidosis, indicating an effect unrelated to the degree of acidosis. Exposure to 10% CO2 increased the burden of P. aeruginosa in the lungs, spleen, and liver, but did not alter lung injury attributable to pneumonia. Hypercapnia did not reduce pulmonary neutrophil recruitment during infection, but alveolar neutrophils from 10% CO2-exposed mice phagocytosed fewer bacteria and produced less H2O2 than neutrophils from air-exposed mice. Secretion of IL-6 and TNF in the lungs of 10% CO2-exposed mice was decreased 7 hours, but not 15 hours, after the onset of pneumonia, indicating that hypercapnia inhibited the early cytokine response to infection. The increase in pneumonia mortality caused by elevated CO2 was reversible when hypercapnic mice were returned to breathing air before or immediately after infection. These results suggest that hypercapnia may increase the susceptibility to and/or worsen the outcome of lung infections in patients with severe lung disease.


Journal of Biological Chemistry | 2015

High CO2 Levels Cause Skeletal Muscle Atrophy via AMP-activated Kinase (AMPK), FoxO3a Protein, and Muscle-specific Ring Finger Protein 1 (MuRF1)

Ariel Jaitovich; Martín Angulo; Emilia Lecuona; Laura A. Dada; Lynn C. Welch; Yuan Cheng; Galina A. Gusarova; Ermelinda Ceco; Chang Liu; Masahiko Shigemura; Esther Barreiro; Cam Patterson; Gustavo A. Nader; Jacob I. Sznajder

Background: CO2 retention and skeletal muscle atrophy occur in patients with lung diseases and are associated with poor clinical outcomes. Results: Hypercapnia leads to AMPK/FoxO3a/MuRF1-dependent muscle fiber size reduction. Conclusion: Hypercapnia activates a signaling pathway leading to skeletal muscle atrophy. Significance: High CO2 levels directly activate a proteolytic program of skeletal muscle atrophy which is of relevance to patients with lung diseases. Patients with chronic obstructive pulmonary disease, acute lung injury, and critical care illness may develop hypercapnia. Many of these patients often have muscle dysfunction which increases morbidity and impairs their quality of life. Here, we investigated whether hypercapnia leads to skeletal muscle atrophy. Mice exposed to high CO2 had decreased skeletal muscle wet weight, fiber diameter, and strength. Cultured myotubes exposed to high CO2 had reduced fiber diameter, protein/DNA ratios, and anabolic capacity. High CO2 induced the expression of MuRF1 in vivo and in vitro, whereas MuRF1−/− mice exposed to high CO2 did not develop muscle atrophy. AMP-activated kinase (AMPK), a metabolic sensor, was activated in myotubes exposed to high CO2, and loss-of-function studies showed that the AMPKα2 isoform is necessary for muscle-specific ring finger protein 1 (MuRF1) up-regulation and myofiber size reduction. High CO2 induced AMPKα2 activation, triggering the phosphorylation and nuclear translocation of FoxO3a, and leading to an increase in MuRF1 expression and myotube atrophy. Accordingly, we provide evidence that high CO2 activates skeletal muscle atrophy via AMPKα2-FoxO3a-MuRF1, which is of biological and potentially clinical significance in patients with lung diseases and hypercapnia.


FEBS Letters | 2010

Extracellular signal-regulated kinase (ERK) participates in the hypercapnia-induced Na,K-ATPase downregulation.

Lynn C. Welch; Emilia Lecuona; Arturo Briva; Humberto E. Trejo; Laura A. Dada; Jacob I. Sznajder

Hypercapnia has been shown to impair alveolar fluid reabsorption (AFR) by decreasing Na,K‐ATPase activity. Extracellular signal‐regulated kinase pathway (ERK) is activated under conditions of cellular stress and has been known to regulate the Na,K‐ATPase. Here, we show that hypercapnia leads to ERK activation in a time‐dependent manner in alveolar epithelial cells (AEC). Inhibition of ERK by U0126 or siRNA prevented both the hypercapnia‐induced Na,K‐ATPase endocytosis and impairment of AFR. Moreover, ERK inhibition prevented AMPK activation, a known modulator of hypercapnia‐induced Na,K‐ATPase endocytosis. Accordingly, these data suggest that hypercapnia‐induced Na,K‐ATPase endocytosis is dependent on ERK activation in AEC and that ERK plays an important role in hypercapnia‐induced impairment of AFR in rat lungs.


Journal of Cell Science | 2009

Myosin-Va restrains the trafficking of Na+/K+-ATPase-containing vesicles in alveolar epithelial cells

Emilia Lecuona; Alexander A. Minin; Humberto E. Trejo; Jiwang Chen; Alejandro P. Comellas; Haiying Sun; Doris Grillo; Oxana Nekrasova; Lynn C. Welch; Igal Szleifer; Vladimir I. Gelfand; Jacob I. Sznajder

Stimulation of Na+/K+-ATPase activity in alveolar epithelial cells by cAMP involves its recruitment from intracellular compartments to the plasma membrane. Here, we studied the role of the actin molecular motor myosin-V in this process. We provide evidence that, in alveolar epithelial cells, cAMP promotes Na+/K+-ATPase recruitment to the plasma membrane by increasing the average speed of Na+/K+-ATPase-containing vesicles moving to the cell periphery. We found that three isoforms of myosin-V are expressed in alveolar epithelial cells; however, only myosin-Va and Vc colocalized with the Na+/K+-ATPase in intracellular membrane fractions. Overexpression of dominant-negative myosin-Va or knockdown with specific shRNA increased the average speed and distance traveled by the Na+/K+-ATPase-containing vesicles, as well as the Na+/K+-ATPase activity and protein abundance at the plasma membrane to similar levels as those observed with cAMP stimulation. These data show that myosin-Va has a role in restraining Na+/K+-ATPase-containing vesicles within intracellular pools and that this restrain is released after stimulation by cAMP allowing the recruitment of the Na+/K+-ATPase to the plasma membrane and thus increased activity.


PLOS ONE | 2012

Evolutionary Conserved Role of c-Jun-N-Terminal Kinase in CO2-Induced Epithelial Dysfunction

István Vadász; Laura A. Dada; Arturo Briva; Iiro Taneli Helenius; Kfir Sharabi; Lynn C. Welch; Aileen M. Kelly; Benno Grzesik; G. R. Scott Budinger; Jing Liu; Werner Seeger; Greg J. Beitel; Yosef Gruenbaum; Jacob I. Sznajder

Elevated CO2 levels (hypercapnia) occur in patients with respiratory diseases and impair alveolar epithelial integrity, in part, by inhibiting Na,K-ATPase function. Here, we examined the role of c-Jun N-terminal kinase (JNK) in CO2 signaling in mammalian alveolar epithelial cells as well as in diptera, nematodes and rodent lungs. In alveolar epithelial cells, elevated CO2 levels rapidly induced activation of JNK leading to downregulation of Na,K-ATPase and alveolar epithelial dysfunction. Hypercapnia-induced activation of JNK required AMP-activated protein kinase (AMPK) and protein kinase C-ζ leading to subsequent phosphorylation of JNK at Ser-129. Importantly, elevated CO2 levels also caused a rapid and prominent activation of JNK in Drosophila S2 cells and in C. elegans. Paralleling the results with mammalian epithelial cells, RNAi against Drosophila JNK fully prevented CO2-induced downregulation of Na,K-ATPase in Drosophila S2 cells. The importance and specificity of JNK CO2 signaling was additionally demonstrated by the ability of mutations in the C. elegans JNK homologs, jnk-1 and kgb-2 to partially rescue the hypercapnia-induced fertility defects but not the pharyngeal pumping defects. Together, these data provide evidence that deleterious effects of hypercapnia are mediated by JNK which plays an evolutionary conserved, specific role in CO2 signaling in mammals, diptera and nematodes.

Collaboration


Dive into the Lynn C. Welch's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiwang Chen

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Arturo Briva

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yochai Adir

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Zaher S. Azzam

Technion – Israel Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge