1
|
Monaco A, Ovryn B, Axis J, Amsler K. The Epithelial Cell Leak Pathway. Int J Mol Sci 2021; 22:ijms22147677. [PMID: 34299297 PMCID: PMC8305272 DOI: 10.3390/ijms22147677] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023] Open
Abstract
The epithelial cell tight junction structure is the site of the transepithelial movement of solutes and water between epithelial cells (paracellular permeability). Paracellular permeability can be divided into two distinct pathways, the Pore Pathway mediating the movement of small ions and solutes and the Leak Pathway mediating the movement of large solutes. Claudin proteins form the basic paracellular permeability barrier and mediate the movement of small ions and solutes via the Pore Pathway. The Leak Pathway remains less understood. Several proteins have been implicated in mediating the Leak Pathway, including occludin, ZO proteins, tricellulin, and actin filaments, but the proteins comprising the Leak Pathway remain unresolved. Many aspects of the Leak Pathway, such as its molecular mechanism, its properties, and its regulation, remain controversial. In this review, we provide a historical background to the evolution of the Leak Pathway concept from the initial examinations of paracellular permeability. We then discuss current information about the properties of the Leak Pathway and present current theories for the Leak Pathway. Finally, we discuss some recent research suggesting a possible molecular basis for the Leak Pathway.
Collapse
Affiliation(s)
- Ashley Monaco
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Boulevard, Old Westbury, NY 11568, USA; (A.M.); (J.A.)
| | - Ben Ovryn
- Department of Physics, New York Institute of Technology, Northern Boulevard, Old Westbury, NY 11568, USA;
| | - Josephine Axis
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Boulevard, Old Westbury, NY 11568, USA; (A.M.); (J.A.)
| | - Kurt Amsler
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Boulevard, Old Westbury, NY 11568, USA; (A.M.); (J.A.)
- Correspondence: ; Tel.: +1-516-686-3716
| |
Collapse
|
2
|
Yan J, Tang Y, Zhong X, Huang H, Wei H, Jin Y, He Y, Cao J, Jin L, Hu B. ROCK inhibitor attenuates carbon blacks-induced pulmonary fibrosis in mice via Rho/ROCK/NF-kappa B pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:1476-1484. [PMID: 33792148 DOI: 10.1002/tox.23135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/13/2021] [Indexed: 06/12/2023]
Abstract
Exposure to carbon blacks (CBs) has been associated with the progression of pulmonary fibrosis, whereas the mechanism is still not clear. We therefore aimed to investigate the effect of RhoA/ROCK pathway on pulmonary fibrosis caused by CBs exposure. Western blot analysis indicated that CBs could promote the activation of RhoA/ROCK pathway and phosphorylation of p65 and IκBα in mice lung. However, ROCK inhibitor Y-27632 could attenuate phosphorylation levels of p65 and IκBα and restore histopathological changes of the lung tissue. Then, we evaluated the effect of RhoA/ROCK pathway on pulmonary fibrosis by detecting the expression levels of α-SMA, vimentin, and Collagen type-I (Col-I), which could be partly inhibited by Y-27632. It was assumed that inhibition of ROCK could be a promising therapeutic candidate for CBs-induced pulmonary fibrosis, which possibly through the blockage of RhoA/ROCK/NF-κB pathway.
Collapse
Affiliation(s)
- Junyan Yan
- School of Life Science, Shaoxing University, Shaoxing, China
| | - Yaxin Tang
- School of Life Science, Shaoxing University, Shaoxing, China
| | - Xin Zhong
- School of Life Science, Shaoxing University, Shaoxing, China
| | - Huarong Huang
- College of Life and Environmental Science, Hangzhou Normal University, Hangzhou, China
| | - Haonan Wei
- School of Life Science, Shaoxing University, Shaoxing, China
| | - Yulei Jin
- School of Life Science, Shaoxing University, Shaoxing, China
| | - Yanjiang He
- School of Life Science, Shaoxing University, Shaoxing, China
| | - Jinqiao Cao
- School of Life Science, Shaoxing University, Shaoxing, China
| | - Lifang Jin
- School of Life Science, Shaoxing University, Shaoxing, China
- Shaoxing Academy of Biomedicine of Zhejiang Sci-Tech University, Zhejiang, China
| | - Baowei Hu
- School of Life Science, Shaoxing University, Shaoxing, China
| |
Collapse
|
3
|
Kim J, Tamura A, Tsukita S, Park S. Uniaxial stretching device for studying maturity-dependent morphological response of epithelial cell monolayers to tensile strain. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2021.04.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
4
|
Varadarajan S, Stephenson RE, Miller AL. Multiscale dynamics of tight junction remodeling. J Cell Sci 2019; 132:132/22/jcs229286. [PMID: 31754042 DOI: 10.1242/jcs.229286] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Epithelial cells form tissues that generate biological barriers in the body. Tight junctions (TJs) are responsible for maintaining a selectively permeable seal between epithelial cells, but little is known about how TJs dynamically remodel in response to physiological forces that challenge epithelial barrier function, such as cell shape changes (e.g. during cell division) or tissue stretching (e.g. during developmental morphogenesis). In this Review, we first introduce a framework to think about TJ remodeling across multiple scales: from molecular dynamics, to strand dynamics, to cell- and tissue-scale dynamics. We then relate knowledge gained from global perturbations of TJs to emerging information about local TJ remodeling events, where transient localized Rho activation and actomyosin-mediated contraction promote TJ remodeling to repair local leaks in barrier function. We conclude by identifying emerging areas in the field and propose ideas for future studies that address unanswered questions about the mechanisms that drive TJ remodeling.
Collapse
Affiliation(s)
- Saranyaraajan Varadarajan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Rachel E Stephenson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Ann L Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, United States
| |
Collapse
|
5
|
Ferreira CHF, Shifrin Y, Pan J, Ivanovska J, McNamara PJ, Belik J. The newborn rat gastric emptying rate is volume and not developmentally dependent. Neurogastroenterol Motil 2018; 30:e13233. [PMID: 29024213 DOI: 10.1111/nmo.13233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gastric residuals are a common finding in enterally fed preterm neonates and traditionally thought to reflect immaturity-related delayed gastric emptying. Adult human data suggest that the meal volume regulate the gastric emptying rate, but early in life, this has not been adequately evaluated. The goal of this study was to study the rat postnatal changes in gastric emptying rate and the strain-induced effect on muscle contraction. We hypothesized that the stomach content volume and not developmental factors determines the newborn gastric emptying rate, via the Rho-kinase 2 (ROCK-2) pathway. METHODS Gastric volume and emptying rate measurements were obtained by ultrasound at different postprandial times and the wall strain-dependent changes in muscle contraction were evaluated ex vivo. KEY RESULTS The newborn rat gastric emptying rate was unrelated to postnatal age, maximal 30 min postprandial, and directly proportional to content volume. In vitro measurements showed that the agonist-induced gastric muscle contraction was directly proportional to the stomach wall strain. These changes were mediated via upregulation of ROCK-2 activity. CONCLUSIONS & INFERENCES The newborn rat gastric emptying rate is not developmentally regulated, but dependent on the content volume via wall strain-induced ROCK-2 activation. Further clinical studies addressing the content volume effect on the rate of gastric emptying are warranted, to enhance feeding tolerance in preterm neonates.
Collapse
Affiliation(s)
- C H F Ferreira
- Hospital das Clinicas, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Y Shifrin
- Departments of Paediatrics and Physiology, Translational Medicine Program, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - J Pan
- Departments of Paediatrics and Physiology, Translational Medicine Program, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - J Ivanovska
- Departments of Paediatrics and Physiology, Translational Medicine Program, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - P J McNamara
- Departments of Paediatrics and Physiology, Translational Medicine Program, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - J Belik
- Departments of Paediatrics and Physiology, Translational Medicine Program, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
6
|
Cohen TS. Role of MicroRNA in the Lung's Innate Immune Response. J Innate Immun 2016; 9:243-249. [PMID: 27915347 DOI: 10.1159/000452669] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 10/18/2016] [Indexed: 12/22/2022] Open
Abstract
The immune response to respiratory pathogens must be robust enough to defend the host yet properly constrained such that inflammation-induced tissue damage is avoided. MicroRNA (miRNA) are small noncoding RNA which posttranscriptionally influence gene expression. In this review, we discuss recent experimental evidence of the contribution of miRNA to the lung's response to bacterial and viral pathogens.
Collapse
Affiliation(s)
- Taylor S Cohen
- Department of Infectious Disease, Medimmune, Gaithersburg, MD, USA
| |
Collapse
|
7
|
Tian Y, Gawlak G, O'Donnell JJ, Birukova AA, Birukov KG. Activation of Vascular Endothelial Growth Factor (VEGF) Receptor 2 Mediates Endothelial Permeability Caused by Cyclic Stretch. J Biol Chem 2016; 291:10032-45. [PMID: 26884340 DOI: 10.1074/jbc.m115.690487] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Indexed: 01/28/2023] Open
Abstract
High tidal volume mechanical ventilation and the resultant excessive mechanical forces experienced by lung vascular endothelium are known to lead to increased vascular endothelial leak, but the underlying molecular mechanisms remain incompletely understood. One reported mechanotransduction pathway of increased endothelial cell (EC) permeability caused by high magnitude cyclic stretch (18% CS) involves CS-induced activation of the focal adhesion associated signalosome, which triggers Rho GTPase signaling. This study identified an alternative pathway of CS-induced EC permeability. We show here that high magnitude cyclic stretch (18% CS) rapidly activates VEGF receptor 2 (VEGFR2) signaling by dissociating VEGFR2 from VE-cadherin at the cell junctions. This results in VEGFR2 activation, Src-dependent VE-cadherin tyrosine phosphorylation, and internalization leading to increased endothelial permeability. This process is also accompanied by CS-induced phosphorylation and internalization of PECAM1. Importantly, CS-induced endothelial barrier disruption was attenuated by VEGFR2 inhibition. 18% CS-induced EC permeability was linked to dissociation of cell junction scaffold afadin from the adherens junctions. Forced expression of recombinant afadin in pulmonary endothelium attenuated CS-induced VEGFR2 and VE-cadherin phosphorylation, preserved adherens junction integrity and VEGFR2·VE-cadherin complex, and suppressed CS-induced EC permeability. This study shows for the first time a mechanism whereby VEGFR2 activation mediates EC permeability induced by pathologically relevant cyclic stretch. In this mechanism, CS induces dissociation of the VE-cadherin·VEGFR2 complex localized at the adherens juctions, causing activation of VEGFR2, VEGFR2-mediated Src-dependent phosphorylation of VE-cadherin, disassembly of adherens junctions, and EC barrier failure.
Collapse
Affiliation(s)
- Yufeng Tian
- From the Lung Injury Center and Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Grzegorz Gawlak
- From the Lung Injury Center and Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| | - James J O'Donnell
- From the Lung Injury Center and Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Anna A Birukova
- From the Lung Injury Center and Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Konstantin G Birukov
- From the Lung Injury Center and Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
8
|
Fujii Y, Funakoshi T, Unuma K, Noritake K, Aki T, Uemura K. Hydrogen sulfide donor NaHS induces death of alveolar epithelial L2 cells that is associated with cellular shrinkage, transgelin expression and myosin phosphorylation. J Toxicol Sci 2016; 41:645-54. [DOI: 10.2131/jts.41.645] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Yusuke Fujii
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Takeshi Funakoshi
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Kanako Noritake
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| |
Collapse
|
9
|
Prakash YS, Tschumperlin DJ, Stenmark KR. Coming to terms with tissue engineering and regenerative medicine in the lung. Am J Physiol Lung Cell Mol Physiol 2015; 309:L625-38. [PMID: 26254424 DOI: 10.1152/ajplung.00204.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/04/2015] [Indexed: 01/10/2023] Open
Abstract
Lung diseases such as emphysema, interstitial fibrosis, and pulmonary vascular diseases cause significant morbidity and mortality, but despite substantial mechanistic understanding, clinical management options for them are limited, with lung transplantation being implemented at end stages. However, limited donor lung availability, graft rejection, and long-term problems after transplantation are major hurdles to lung transplantation being a panacea. Bioengineering the lung is an exciting and emerging solution that has the ultimate aim of generating lung tissues and organs for transplantation. In this article we capture and review the current state of the art in lung bioengineering, from the multimodal approaches, to creating anatomically appropriate lung scaffolds that can be recellularized to eventually yield functioning, transplant-ready lungs. Strategies for decellularizing mammalian lungs to create scaffolds with native extracellular matrix components vs. de novo generation of scaffolds using biocompatible materials are discussed. Strengths vs. limitations of recellularization using different cell types of various pluripotency such as embryonic, mesenchymal, and induced pluripotent stem cells are highlighted. Current hurdles to guide future research toward achieving the clinical goal of transplantation of a bioengineered lung are discussed.
Collapse
Affiliation(s)
- Y S Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota;
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota; Division of Pulmonary Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| |
Collapse
|
10
|
Song MJ, Davis CI, Lawrence GG, Margulies SS. Local influence of cell viability on stretch-induced permeability of alveolar epithelial cell monolayers. Cell Mol Bioeng 2015; 9:65-72. [PMID: 26958093 DOI: 10.1007/s12195-015-0405-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Ventilator induced lung injury (VILI), often attributed to over-distension of the alveolar epithelial cell layer, can trigger loss of barrier function. Alveolar epithelial cell monolayers can be used as an idealized in vitro model of the pulmonary epithelium, with cell death and tight junction disruption and permeability employed to estimate stretch-induced changes in barrier function. We adapted a method published for vascular endothelial permeability, compare its sensitivity with our previously published method, and determine the relationship between breeches in barrier properties after stretch and regions of cell death After 4-5 days in culture, primary rat alveolar epithelial cells seeded on plasma treated polydimethylsiloxane membrane coated with biotin-labeled fibronectin, or fibronectin alone were stretched in the presence of FITC-tagged streptavidin (biotin-labeled membrane) or BODIPY-ouabain. We found that the FITC-labeling method was a more sensitive indicator of permeability disruption, with significantly larger positively stained areas visible in the presence of stretch and with ATP production inhibitor Antimycin-A. Triple-stained images with Hoescht (nuclei), Ethidium Homodimer (EthD, damaged cell nuclei) and FITC (permeable regions) were used to determine that within permeable regions intact cells were positioned closer to damaged cells than in non-permeable regions. We concluded that local cell death may be an important contributor to barrier integrity.
Collapse
Affiliation(s)
- M J Song
- Department of Bioengineering, University of Pennsylvania 210 South 33rd Street, Philadelphia, PA 19104-6321, USA
| | - C I Davis
- Department of Bioengineering, University of Pennsylvania 210 South 33rd Street, Philadelphia, PA 19104-6321, USA
| | - G G Lawrence
- Department of Bioengineering, University of Pennsylvania 210 South 33rd Street, Philadelphia, PA 19104-6321, USA
| | - S S Margulies
- Department of Bioengineering, University of Pennsylvania 210 South 33rd Street, Philadelphia, PA 19104-6321, USA
| |
Collapse
|
11
|
Ibuprofen protects ventilator-induced lung injury by downregulating Rho-kinase activity in rats. BIOMED RESEARCH INTERNATIONAL 2014; 2014:749097. [PMID: 25019086 PMCID: PMC4075182 DOI: 10.1155/2014/749097] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 05/15/2014] [Accepted: 05/19/2014] [Indexed: 12/20/2022]
Abstract
Background. Ventilator-induced lung injury-(VILI-) induced endothelial permeability is regulated through the Rho-dependent signaling pathway. Ibuprofen inhibits Rho activation in animal models of spinal-cord injury and Alzheimer's disease. The study aims to investigate ibuprofen effects on high tidal volume associated VILI. Methods. Twenty-eight adult male Sprague-Dawley rats were randomized to receive a ventilation strategy with three different interventions for 2 h: (1) a high-volume zero-positive end-expiratory pressure (PEEP) (HVZP) group; (2) an HVZP + ibuprofen 15 mg/kg group; and (3) an HVZP + ibuprofen 30 mg/kg group. A fourth group without ventilation served as the control group. Rho-kinase activity was determined by ratio of phosphorylated ezrin, radixin, and moesin (p-ERM), substrates of Rho-kinase, to total ERM. VILI was characterized by increased pulmonary protein leak, wet-to-dry weight ratio, cytokines level, and Rho guanine nucleotide exchange factor (GEF-H1), RhoA activity, p-ERM/total ERM, and p-myosin light chain (MLC) protein expression. Results. Ibuprofen pretreatment significantly reduced the HVZP ventilation-induced increase in pulmonary protein leak, wet-to-dry weight ratio, bronchoalveolar lavage fluid interleukin-6 and RANTES levels, and lung GEF-H1, RhoA activity, p-ERM/total ERM, and p-MLC protein expression. Conclusion. Ibuprofen attenuated high tidal volume induced pulmonary endothelial hyperpermeability. This protective effect was associated with a reduced Rho-kinase activity.
Collapse
|
12
|
Goncharova EA, Goncharov DA, James ML, Atochina-Vasserman EN, Stepanova V, Hong SB, Li H, Gonzales L, Baba M, Linehan WM, Gow AJ, Margulies S, Guttentag S, Schmidt LS, Krymskaya VP. Folliculin controls lung alveolar enlargement and epithelial cell survival through E-cadherin, LKB1, and AMPK. Cell Rep 2014; 7:412-423. [PMID: 24726356 PMCID: PMC4034569 DOI: 10.1016/j.celrep.2014.03.025] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/30/2014] [Accepted: 03/10/2014] [Indexed: 12/21/2022] Open
Abstract
Spontaneous pneumothoraces due to lung cyst rupture afflict patients with the rare disease Birt-Hogg-Dubé (BHD) syndrome, which is caused by mutations of the tumor suppressor gene folliculin (FLCN). The underlying mechanism of the lung manifestations in BHD is unclear. We show that BHD lungs exhibit increased alveolar epithelial cell apoptosis and that Flcn deletion in mouse lung epithelium leads to cell apoptosis, alveolar enlargement, and an impairment of both epithelial barrier and overall lung function. We find that Flcn-null epithelial cell apoptosis is the result of impaired AMPK activation and increased cleaved caspase-3. AMPK activator LKB1 and E-cadherin are downregulated by Flcn loss and restored by its expression. Correspondingly, Flcn-null cell survival is rescued by the AMPK activator AICAR or constitutively active AMPK. AICAR also improves lung condition of Flcn(f/f):SP-C-Cre mice. Our data suggest that lung cysts in BHD may result from an underlying defect in alveolar epithelial cell survival, attributable to FLCN regulation of the E-cadherin-LKB1-AMPK axis.
Collapse
Affiliation(s)
- Elena A Goncharova
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, Department of Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Dmitry A Goncharov
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, Department of Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Melane L James
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, Department of Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Elena N Atochina-Vasserman
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, Department of Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Victoria Stepanova
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Seung-Beom Hong
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, Department of Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Hua Li
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, Department of Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Linda Gonzales
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Masaya Baba
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - W Marston Linehan
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Andrew J Gow
- Department of Pharmacology & Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | - Susan Margulies
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susan Guttentag
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Laura S Schmidt
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA; Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 20892, USA
| | - Vera P Krymskaya
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, Department of Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
13
|
Gawlak G, Tian Y, O'Donnell JJ, Tian X, Birukova AA, Birukov KG. Paxillin mediates stretch-induced Rho signaling and endothelial permeability via assembly of paxillin-p42/44MAPK-GEF-H1 complex. FASEB J 2014; 28:3249-60. [PMID: 24706358 DOI: 10.1096/fj.13-245142] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Suboptimal ventilator support or regional ventilation heterogeneity in inflamed lungs causes excessive tissue distension, which triggers stretch-induced pathological signaling and may lead to vascular leak and lung dysfunction. Focal adhesions (FAs) are cell-substrate adhesive complexes participating in cellular mechanotransduction and regulation of the Rho GTPase pathway. Stretch-induced Rho regulation remains poorly understood. We used human lung endothelial cells (ECs) exposed to pathological cyclic stretch (CS) at 18% distension to test the hypothesis that FA protein paxillin participates in CS-induced Rho activation by recruiting the Rho-specific guanine nucleotide exchange factor GEF-H1. CS induced phosphorylation of paxillin and activated p42/44-MAP kinase, Rho GTPase, and paxillin/GEF-H1/p42/44-MAPK association. CS caused nearly 2-fold increase in EC permeability, which was attenuated by paxillin knockdown. Expression of the paxillin-Y31/118F phosphorylation mutant decreased the CS-induced paxillin/GEF-H1 association (16.3 ± 4.1%), GEF-H1 activation (28.9 ± 9.2%), and EC permeability (28.7 ± 8.1%) but not CS-induced p42/44-MAPK activation. Inhibition of p42/44-MAPK suppressed CS-induced paxillin/GEF-H1 interactions (15.9 ± 7.9%), GEF-H1 activation (11.7 ± 4.3%), and disruption of EC monolayer. Expression of GEF-H1T678A lacking p42/44-MAPK phosphorylation site attenuated Rho activation (31.2±11.6%). We conclude that MAPK-dependent targeting of GEF-H1 to paxillin is involved in the regulation of CS-induced Rho signaling and EC permeability. This study proposes a novel concept of paxillin-GEF-H1-p42/44-MAPK module as a regulator of pathological mechanotransduction.-Gawlak, G., Tian, Y., O'Donnell, J. J., III, Tian, X., Birukova, A. A., Birukov, K. G. Paxillin mediates stretch-induced Rho signaling and endothelial permeability via assembly of paxillin-p42/44MAPK-GEF-H1 complex.
Collapse
Affiliation(s)
- Grzegorz Gawlak
- Lung Injury Center, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Yufeng Tian
- Lung Injury Center, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - James J O'Donnell
- Lung Injury Center, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Xinyong Tian
- Lung Injury Center, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Anna A Birukova
- Lung Injury Center, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Konstantin G Birukov
- Lung Injury Center, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
14
|
Yeganeh B, Wiechec E, Ande SR, Sharma P, Moghadam AR, Post M, Freed DH, Hashemi M, Shojaei S, Zeki AA, Ghavami S. Targeting the mevalonate cascade as a new therapeutic approach in heart disease, cancer and pulmonary disease. Pharmacol Ther 2014; 143:87-110. [PMID: 24582968 DOI: 10.1016/j.pharmthera.2014.02.007] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/04/2014] [Indexed: 12/21/2022]
Abstract
The cholesterol biosynthesis pathway, also known as the mevalonate (MVA) pathway, is an essential cellular pathway that is involved in diverse cell functions. The enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase (HMGCR) is the rate-limiting step in cholesterol biosynthesis and catalyzes the conversion of HMG-CoA to MVA. Given its role in cholesterol and isoprenoid biosynthesis, the regulation of HMGCR has been intensely investigated. Because all cells require a steady supply of MVA, both the sterol (i.e. cholesterol) and non-sterol (i.e. isoprenoid) products of MVA metabolism exert coordinated feedback regulation on HMGCR through different mechanisms. The proper functioning of HMGCR as the proximal enzyme in the MVA pathway is essential under both normal physiologic conditions and in many diseases given its role in cell cycle pathways and cell proliferation, cholesterol biosynthesis and metabolism, cell cytoskeletal dynamics and stability, cell membrane structure and fluidity, mitochondrial function, proliferation, and cell fate. The blockbuster statin drugs ('statins') directly bind to and inhibit HMGCR, and their use for the past thirty years has revolutionized the treatment of hypercholesterolemia and cardiovascular diseases, in particular coronary heart disease. Initially thought to exert their effects through cholesterol reduction, recent evidence indicates that statins also have pleiotropic immunomodulatory properties independent of cholesterol lowering. In this review we will focus on the therapeutic applications and mechanisms involved in the MVA cascade including Rho GTPase and Rho kinase (ROCK) signaling, statin inhibition of HMGCR, geranylgeranyltransferase (GGTase) inhibition, and farnesyltransferase (FTase) inhibition in cardiovascular disease, pulmonary diseases (e.g. asthma and chronic obstructive pulmonary disease (COPD)), and cancer.
Collapse
Affiliation(s)
- Behzad Yeganeh
- Hospital for Sick Children Research Institute, Department of Physiology & Experimental Medicine, University of Toronto, Toronto, Canada
| | - Emilia Wiechec
- Dept. Clinical & Experimental Medicine, Division of Cell Biology & Integrative Regenerative Med. Center (IGEN), Linköping University, Sweden
| | - Sudharsana R Ande
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan Sharma
- Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, 4C46 HRIC, 3280 Hospital Drive NW, Calgary, Alberta, Canada
| | - Adel Rezaei Moghadam
- Scientific Association of Veterinary Medicine, Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran; Young Researchers and Elite Club, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| | - Martin Post
- Hospital for Sick Children Research Institute, Department of Physiology & Experimental Medicine, University of Toronto, Toronto, Canada
| | - Darren H Freed
- Department of Physiology, St. Boniface Research Centre, University of Manitoba, Winnipeg, Canada
| | - Mohammad Hashemi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Shahla Shojaei
- Department of Biochemistry, Recombinant Protein Laboratory, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir A Zeki
- U.C. Davis, School of Medicine, U.C. Davis Medical Center, Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Comparative Respiratory Biology & Medicine, Davis, CA, USA.
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, St. Boniface Research Centre, Manitoba Institute of Child Health, Biology of Breathing Theme, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
15
|
Wilhelm KR, Roan E, Ghosh MC, Parthasarathi K, Waters CM. Hyperoxia increases the elastic modulus of alveolar epithelial cells through Rho kinase. FEBS J 2013; 281:957-69. [PMID: 24289040 DOI: 10.1111/febs.12661] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 11/05/2013] [Accepted: 11/26/2013] [Indexed: 12/31/2022]
Abstract
Patients with acute lung injury are administered high concentrations of oxygen during mechanical ventilation, and while both hyperoxia and mechanical ventilation are necessary, each can independently cause additional injury. However, the precise mechanisms that lead to injury are not well understood. We hypothesized that alveolar epithelial cells may be more susceptible to injury caused by mechanical ventilation because hyperoxia causes cells to be stiffer due to increased filamentous actin (f-actin) formation via the GTPase RhoA and its effecter Rho kinase (ROCK). We examined cytoskeletal structures in cultured murine lung alveolar epithelial cells (MLE-12) under normoxic and hyperoxic (48 h) conditions. We also measured cell elasticity (E) using an atomic force microscope in the indenter mode. Hyperoxia caused increased f-actin stress fibers and bundle formation, an increase in g- and f-actin, an increase in nuclear area and a decrease in nuclear height, and cells became stiffer (higher E). Treatment with an inhibitor (Y-27632) of ROCK significantly decreased E and prevented the cytoskeletal changes, while it did not influence the nuclear height and area. Pre-exposure of cells to hyperoxia promoted detachment when cells were subsequently stretched cyclically, but the ROCK inhibitor prevented this effect. Hyperoxia caused thickening of vinculin focal adhesion plaques, and inhibition of ROCK reduced the formation of distinct focal adhesion plaques. Phosphorylation of focal adhesion kinase was significantly reduced by both hyperoxia and treatment with Y-27632. Hyperoxia caused increased cell stiffness and promoted cell detachment during stretch. These effects were ameliorated by inhibition of ROCK.
Collapse
Affiliation(s)
- Kristina R Wilhelm
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | | | | |
Collapse
|
16
|
Herold S, Gabrielli NM, Vadász I. Novel concepts of acute lung injury and alveolar-capillary barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 2013; 305:L665-81. [PMID: 24039257 DOI: 10.1152/ajplung.00232.2013] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this review we summarize recent major advances in our understanding on the molecular mechanisms, mediators, and biomarkers of acute lung injury (ALI) and alveolar-capillary barrier dysfunction, highlighting the role of immune cells, inflammatory and noninflammatory signaling events, mechanical noxae, and the affected cellular and molecular entities and functions. Furthermore, we address novel aspects of resolution and repair of ALI, as well as putative candidates for treatment of ALI, including pharmacological and cellular therapeutic means.
Collapse
Affiliation(s)
- Susanne Herold
- Dept. of Internal Medicine, Justus Liebig Univ., Universities of Giessen and Marburg Lung Center, Klinikstrasse 33, 35392 Giessen, Germany.
| | | | | |
Collapse
|
17
|
Davidovich N, DiPaolo BC, Lawrence GG, Chhour P, Yehya N, Margulies SS. Cyclic stretch-induced oxidative stress increases pulmonary alveolar epithelial permeability. Am J Respir Cell Mol Biol 2013; 49:156-64. [PMID: 23526210 DOI: 10.1165/rcmb.2012-0252oc] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mechanical ventilation with high tidal volumes has been associated with pulmonary alveolar flooding. Understanding the mechanisms underlying cyclic stretch-induced increases in alveolar epithelial permeability may be important in designing preventive measures for acute lung injury. In this work, we assessed whether cyclic stretch leads to the generation of reactive oxygen species in type I-like alveolar epithelial cells, which increase monolayer permeability via activation of NF-κB and extracellular signal-regulated kinase (ERK). We cyclically stretched type I-like rat primary alveolar epithelial cells at magnitudes of 12, 25, and 37% change in surface area (ΔSA) for 10 to 120 minutes. High levels of reactive oxygen species and of superoxide and NO specifically were detected in cells stretched at 37% ΔSA for 10 to 120 minutes. Exogenous superoxide and NO stimulation increased epithelial permeability in unstretched cells, which was preventable by the NF-κB inhibitor MG132. The cyclic stretch-induced increase in permeability was decreased by the superoxide scavenger tiron and by MG132. Furthermore, tiron had a dramatic protective effect on in vivo lung permeability under mechanical ventilation conditions. Cyclic stretch increased the activation of the NF-κB signaling pathway, which was significantly decreased with the ERK inhibitor U0126. Altogether, our in vitro and in vivo data demonstrate the sensitivity of permeability to stretch- and ventilation-induced superoxide production, suggesting that using antioxidants may be helpful in the prevention and treatment of ventilator-induced lung injury.
Collapse
Affiliation(s)
- Nurit Davidovich
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
18
|
Hussein O, Walters B, Stroetz R, Valencia P, McCall D, Hubmayr RD. Biophysical determinants of alveolar epithelial plasma membrane wounding associated with mechanical ventilation. Am J Physiol Lung Cell Mol Physiol 2013; 305:L478-84. [PMID: 23997173 DOI: 10.1152/ajplung.00437.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Mechanical ventilation may cause harm by straining lungs at a time they are particularly prone to injury from deforming stress. The objective of this study was to define the relative contributions of alveolar overdistension and cyclic recruitment and "collapse" of unstable lung units to membrane wounding of alveolar epithelial cells. We measured the interactive effects of tidal volume (VT), transpulmonary pressure (PTP), and of airspace liquid on the number of alveolar epithelial cells with plasma membrane wounds in ex vivo mechanically ventilated rat lungs. Plasma membrane integrity was assessed by propidium iodide (PI) exclusion in confocal images of subpleural alveoli. Cyclic inflations of normal lungs from zero end-expiratory pressure to 40 cmH2O produced VT values of 56.9 ± 3.1 ml/kg and were associated with 0.12 ± 0.12 PI-positive cells/alveolus. A preceding tracheal instillation of normal saline (3 ml) reduced VT to 49.1 ± 6 ml/kg but was associated with a significantly greater number of wounded alveolar epithelial cells (0.52 ± 0.16 cells/alveolus; P < 0.01). Mechanical ventilation of completely saline-filled lungs with saline (VT = 52 ml/kg) to pressures between 10 and 15 cmH2O was associated with the least number of wounded epithelial cells (0.02 ± 0.02 cells/alveolus; P < 0.01). In mechanically ventilated, partially saline-filled lungs, the number of wounded cells increased substantially with VT, but, once VT was accounted for, wounding was independent of maximal PTP. We found that interfacial stress associated with the generation and destruction of liquid bridges in airspaces is the primary biophysical cell injury mechanism in mechanically ventilated lungs.
Collapse
Affiliation(s)
- Omar Hussein
- Mayo Clinic, Division of Pulmonary and Critical Care Medicine, 200 1st St. SW, Rochester, MN 55905.
| | | | | | | | | | | |
Collapse
|
19
|
Dipaolo BC, Davidovich N, Kazanietz MG, Margulies SS. Rac1 pathway mediates stretch response in pulmonary alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 2013; 305:L141-53. [PMID: 23686855 DOI: 10.1152/ajplung.00298.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alveolar epithelial cells (AECs) maintain the pulmonary blood-gas barrier integrity with gasketlike intercellular tight junctions (TJ) that are anchored internally to the actin cytoskeleton. We have previously shown that AEC monolayers stretched cyclically and equibiaxially undergo rapid magnitude- and frequency-dependent actin cytoskeletal remodeling to form perijunctional actin rings (PJARs). In this work, we show that even 10 min of stretch induced increases in the phosphorylation of Akt and LIM kinase (LIMK) and decreases in cofilin phosphorylation, suggesting that the Rac1/Akt pathway is involved in these stretch-mediated changes. We confirmed that Rac1 inhibitors wortmannin or EHT-1864 decrease stretch-stimulated Akt and LIMK phosphorylation and that Rac1 agonists PIP3 or PDGF increase phosphorylation of these proteins in unstretched cells. We also confirmed that Rac1 pathway inhibition during stretch modulated stretch-induced changes in occludin content and monolayer permeability, actin remodeling and PJAR formation, and cell death. As further validation, overexpression of Rac GTPase-activating protein β2-chimerin also preserved monolayer barrier properties in stretched monolayers. In summary, our data suggest that constitutive activity of Rac1, which is necessary for stretch-induced activation of the Rac1 downstream proteins, mediates stretch-induced increases in permeability and PJAR formation.
Collapse
Affiliation(s)
- Brian C Dipaolo
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
20
|
Cohen TS, DiPaolo BC, Gray Lawrence G, Margulies SS. Sepsis enhances epithelial permeability with stretch in an actin dependent manner. PLoS One 2012; 7:e38748. [PMID: 22723883 PMCID: PMC3378620 DOI: 10.1371/journal.pone.0038748] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/11/2012] [Indexed: 01/11/2023] Open
Abstract
Ventilation of septic patients often leads to the development of edema and impaired gas exchange. We hypothesized that septic alveolar epithelial monolayers would experience stretch-induced barrier dysfunction at a lower magnitude of stretch than healthy alveolar epithelial monolayers. Alveolar epithelial cells were isolated from rats 24 hours after cecal ligation and double puncture (2CLP) or sham surgery. Following a 5-day culture period, monolayers were cyclically stretched for 0, 10, or 60 minutes to a magnitude of 12% or 25% change in surface area (ΔSA). Barrier function, MAPk and myosin light chain (MLC) phosphorylation, tight junction (TJ) protein expression and actin cytoskeletal organization were examined after stretch. Significant increases in epithelial permeability were observed only in 2CLP monolayers at the 12% ΔSA stretch level, and in both 2CLP and sham monolayers at the 25% ΔSA stretch level. Increased permeability in 2CLP monolayers was not associated with MAPk signaling or alterations in expression of TJ proteins. 2CLP monolayers had fewer actin stress fibers before stretch, a more robust stretch-induced actin redistribution, and reduced phosphorylated MLCK than sham monolayers. Jasplakinolide stabilization of the actin cytoskeleton in 2CLP monolayers prevented significant increases in permeability following 60 minutes of stretch to 12% ΔSA. We concluded that septic alveolar epithelial monolayers are more susceptible to stretch-induced barrier dysfunction than healthy monolayers due to actin reorganization.
Collapse
Affiliation(s)
- Taylor S. Cohen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Brian C. DiPaolo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gladys Gray Lawrence
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Susan S. Margulies
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|