1
|
Boelman MB, Hansen TVO, Smith MN, Hammer-Hansen S, Christensen AH, Diness BR. Aortic dissection in a young male with persistent ductus arteriosus and a novel variant in MYLK. Am J Med Genet A 2024; 194:e63458. [PMID: 37921548 DOI: 10.1002/ajmg.a.63458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/29/2023] [Accepted: 10/16/2023] [Indexed: 11/04/2023]
Abstract
Pathogenic variants in several genes involved in the function or regulation of smooth muscle cells (SMC) are known to predispose to congenital heart disease and thoracic aortic aneurysm and dissection (TAAD). Variants in MYLK are primarily known to predispose to TAAD, but a growing body of evidence points toward MYLK also playing an essential role in the regulation of SMC contraction outside the aorta. In this case report, we present a patient with co-occurrence of persistent ductus arteriosus (PDA) and thoracic aortic dissection. Genetic analyses revealed a novel splice acceptor variant (c.3986-1G > A) in MYLK, which segregated with disease in the family. RNA-analyses on fibroblasts showed that the variant induced skipping of exon 24, which resulted in an in-frame deletion of 101 amino acids. These findings suggest that MYLK-associated disease could include a broader phenotypic spectrum than isolated TAAD, including PDA and obstructive pulmonary disease. Genetic analyses could be considered in families with TAAD and PDA or obstructive pulmonary disease.
Collapse
Affiliation(s)
| | - Thomas van Overeem Hansen
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Birgitte Rode Diness
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Kempf CL, Sammani S, Bermudez T, Song JH, Hernon VR, Hufford MK, Burt J, Camp SM, Dudek SM, Garcia JG. Critical Role for the Lung Endothelial Non‐Muscle Myosin Light Chain Kinase Isoform in the Severity of Inflammatory Murine Lung Injury. Pulm Circ 2022; 12:e12061. [PMID: 35514774 PMCID: PMC9063969 DOI: 10.1002/pul2.12061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Carrie L. Kempf
- Department of Medicine University of Arizona Health Sciences Tucson AZ USA
| | - Saad Sammani
- Department of Medicine University of Arizona Health Sciences Tucson AZ USA
| | - Tadeo Bermudez
- Department of Medicine University of Arizona Health Sciences Tucson AZ USA
| | - Jin H. Song
- Department of Medicine University of Arizona Health Sciences Tucson AZ USA
| | | | - Matthew K. Hufford
- Department of Medicine University of Arizona Health Sciences Tucson AZ USA
| | - Jessica Burt
- Department of Medicine University of Arizona Health Sciences Tucson AZ USA
| | - Sara M. Camp
- Department of Medicine University of Arizona Health Sciences Tucson AZ USA
| | - Steven M. Dudek
- Department of Medicine University of Illinois at Chicago Chicago IL USA
| | - Joe G.N. Garcia
- Department of Medicine University of Arizona Health Sciences Tucson AZ USA
| |
Collapse
|
3
|
Sun X, Sun B, Sammani S, Bermudez T, Dudek S, Camp S, Garcia J. Genetic and epigenetic regulation of the non-muscle myosin light chain kinase isoform by lung inflammatory factors and mechanical stress. Clin Sci (Lond) 2021; 135:963-977. [PMID: 33792658 PMCID: PMC8047480 DOI: 10.1042/cs20201448] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/19/2021] [Accepted: 04/01/2021] [Indexed: 12/24/2022]
Abstract
RATIONALE The myosin light chain kinase gene, MYLK, encodes three proteins via unique promoters, including the non-muscle isoform of myosin light chain kinase (nmMLCK), a cytoskeletal protein centrally involved in regulation of vascular integrity. As MYLK coding SNPs are associated with severe inflammatory disorders (asthma, acute respiratory distress syndrome (ARDS)), we explored clinically relevant inflammatory stimuli and promoter SNPs in nmMLCK promoter regulation. METHODS Full-length or serially deleted MYLK luciferase reporter promoter activities were measured in human lung endothelial cells (ECs). SNP-containing non-muscle MYLK (nmMYLK) DNA fragments were generated and nmMYLK promoter binding by transcription factors (TFs) detected by protein-DNA electrophoretic mobility shift assay (EMSA). Promoter demethylation was evaluated by 5-aza-2'-deoxycytidine (5-Aza). A preclinical mouse model of lipopolysaccharide (LPS)-induced acute lung injury (ALI) was utilized for nmMLCK validation. RESULTS Lung EC levels of nmMLCK were significantly increased in LPS-challenged mice and LPS, tumor necrosis factor-α (TNF-α), 18% cyclic stretch (CS) and 5-Aza each significantly up-regulated EC nmMYLK promoter activities. EC exposure to FG-4592, a prolyl hydroxylase inhibitor that increases hypoxia-inducible factor (HIF) expression, increased nmMYLK promoter activity, confirmed by HIF1α/HIF2α silencing. nmMYLK promoter deletion studies identified distal inhibitory and proximal enhancing promoter regions as well as mechanical stretch-, LPS- and TNFα-inducible regions. Insertion of ARDS-associated SNPs (rs2700408, rs11714297) significantly increased nmMYLK promoter activity via increased transcription binding (glial cells missing homolog 1 (GCM1) and intestine-specific homeobox (ISX), respectively). Finally, the MYLK rs78755744 SNP (-261G/A), residing within a nmMYLK CpG island, significantly attenuated 5-Aza-induced promoter activity. CONCLUSION These findings indicate nmMYLK transcriptional regulation by clinically relevant inflammatory factors and ARDS-associated nmMYLK promoter variants are consistent with nmMLCK as a therapeutic target in severe inflammatory disorders.
Collapse
Affiliation(s)
- Xiaoguang Sun
- Department of Medicine, University of Arizona, Tucson, AZ, U.S.A
| | - Belinda L. Sun
- Department of Pathology, University of Arizona, Tucson, AZ, U.S.A
| | - Saad Sammani
- Department of Medicine, University of Arizona, Tucson, AZ, U.S.A
| | - Tadeo Bermudez
- Department of Medicine, University of Arizona, Tucson, AZ, U.S.A
| | - Steven M. Dudek
- Department of Medicine, University of Illinois Chicago, Chicago, IL, U.S.A
| | - Sara M. Camp
- Department of Medicine, University of Arizona, Tucson, AZ, U.S.A
| | - Joe G.N. Garcia
- Department of Medicine, University of Arizona, Tucson, AZ, U.S.A
| |
Collapse
|
4
|
Lynn H, Sun X, Casanova N, Gonzales-Garay M, Bime C, Garcia JGN. Genomic and Genetic Approaches to Deciphering Acute Respiratory Distress Syndrome Risk and Mortality. Antioxid Redox Signal 2019; 31:1027-1052. [PMID: 31016989 PMCID: PMC6939590 DOI: 10.1089/ars.2018.7701] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Acute respiratory distress syndrome (ARDS) is a severe, highly heterogeneous critical illness with staggering mortality that is influenced by environmental factors, such as mechanical ventilation, and genetic factors. Significant unmet needs in ARDS are addressing the paucity of validated predictive biomarkers for ARDS risk and susceptibility that hamper the conduct of successful clinical trials in ARDS and the complete absence of novel disease-modifying therapeutic strategies. Recent Advances: The current ARDS definition relies on clinical characteristics that fail to capture the diversity of disease pathology, severity, and mortality risk. We undertook a comprehensive survey of the available ARDS literature to identify genes and genetic variants (candidate gene and limited genome-wide association study approaches) implicated in susceptibility to developing ARDS in hopes of uncovering novel biomarkers for ARDS risk and mortality and potentially novel therapeutic targets in ARDS. We further attempted to address the well-known health disparities that exist in susceptibility to and mortality from ARDS. Critical Issues: Bioinformatic analyses identified 201 ARDS candidate genes with pathway analysis indicating a strong predominance in key evolutionarily conserved inflammatory pathways, including reactive oxygen species, innate immunity-related inflammation, and endothelial vascular signaling pathways. Future Directions: Future studies employing a system biology approach that combines clinical characteristics, genomics, transcriptomics, and proteomics may allow for a better definition of biologically relevant pathways and genotype-phenotype connections and result in improved strategies for the sub-phenotyping of diverse ARDS patients via molecular signatures. These efforts should facilitate the potential for successful clinical trials in ARDS and yield a better fundamental understanding of ARDS pathobiology.
Collapse
Affiliation(s)
- Heather Lynn
- Department of Physiological Sciences and University of Arizona, Tucson, Arizona.,Department of Health Sciences, University of Arizona, Tucson, Arizona
| | - Xiaoguang Sun
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| | - Nancy Casanova
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| | | | - Christian Bime
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| | - Joe G N Garcia
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| |
Collapse
|
5
|
Evasovic JM, Singer CA. Regulation of IL-17A and implications for TGF-β1 comodulation of airway smooth muscle remodeling in severe asthma. Am J Physiol Lung Cell Mol Physiol 2019; 316:L843-L868. [PMID: 30810068 PMCID: PMC6589583 DOI: 10.1152/ajplung.00416.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/04/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
Severe asthma develops as a result of heightened, persistent symptoms that generally coincide with pronounced neutrophilic airway inflammation. In individuals with severe asthma, symptoms are poorly controlled by high-dose inhaled glucocorticoids and often lead to elevated morbidity and mortality rates that underscore the necessity for novel drug target identification that overcomes limitations in disease management. Many incidences of severe asthma are mechanistically associated with T helper 17 (TH17) cell-derived cytokines and immune factors that mediate neutrophilic influx to the airways. TH17-secreted interleukin-17A (IL-17A) is an independent risk factor for severe asthma that impacts airway smooth muscle (ASM) remodeling. TH17-derived cytokines and diverse immune mediators further interact with structural cells of the airway to induce pathophysiological processes that impact ASM functionality. Transforming growth factor-β1 (TGF-β1) is a pivotal mediator involved in airway remodeling that correlates with enhanced TH17 activity in individuals with severe asthma and is essential to TH17 differentiation and IL-17A production. IL-17A can also reciprocally enhance activation of TGF-β1 signaling pathways, whereas combined TH1/TH17 or TH2/TH17 immune responses may additively impact asthma severity. This review seeks to provide a comprehensive summary of cytokine-driven T cell fate determination and TH17-mediated airway inflammation. It will further review the evidence demonstrating the extent to which IL-17A interacts with various immune factors, specifically TGF-β1, to contribute to ASM remodeling and altered function in TH17-driven endotypes of severe asthma.
Collapse
Affiliation(s)
- Jon M Evasovic
- Department of Pharmacology, School of Medicine, University of Nevada , Reno, Nevada
| | - Cherie A Singer
- Department of Pharmacology, School of Medicine, University of Nevada , Reno, Nevada
| |
Collapse
|
6
|
Single nucleotide polymorphisms in the MYLKP1 pseudogene are associated with increased colon cancer risk in African Americans. PLoS One 2018; 13:e0200916. [PMID: 30161129 PMCID: PMC6116948 DOI: 10.1371/journal.pone.0200916] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/04/2018] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Pseudogenes are paralogues of functional genes historically viewed as defunct due to either the lack of regulatory elements or the presence of frameshift mutations. Recent evidence, however, suggests that pseudogenes may regulate gene expression, although the functional role of pseudogenes remains largely unknown. We previously reported that MYLKP1, the pseudogene of MYLK that encodes myosin light chain kinase (MLCK), is highly expressed in lung and colon cancer cell lines and tissues but not in normal lung or colon. The MYLKP1 promoter is minimally active in normal bronchial epithelial cells but highly active in lung adenocarcinoma cells. In this study, we further validate MYLKP1 as an oncogene via elucidation of the functional role of MYLKP1 genetic variants in colon cancer risk. METHODS Proliferation and migration assays were performed in MYLKP1-transfected colon and lung cancer cell lines (H441, A549) and commercially-available normal lung and colon cells. Fourteen MYLKP1 SNPs (MAFs >0.01) residing within the 4 kb MYLKP1 promoter region, the core 1.4 kb of MYLKP1 gene, and a 4 kb enhancer region were selected and genotyped in a colorectal cancer cohort. MYLKP1 SNP influences on activity of MYLKP1 promoter (2kb) was assessed by dual luciferase reporter assay. RESULTS Cancer cell lines, H441 and A549, exhibited increased MYLKP1 expression, increased MYLKP1 luciferase promoter activity, increased proliferation and migration. Genotyping studies identified two MYLKP1 SNPs (rs12490683; rs12497343) that significantly increase risk of colon cancer in African Americans compared to African American controls. Rs12490683 and rs12497343 further increase MYLKP1 promoter activity compared to the wild type MYLKP1 promoter. CONCLUSION MYLKP1 is a cancer-promoting pseudogene whose genetic variants differentially enhance cancer risk in African American populations.
Collapse
|
7
|
Wirshing ACE, Cram EJ. Spectrin regulates cell contractility through production and maintenance of actin bundles in the Caenorhabditis elegans spermatheca. Mol Biol Cell 2018; 29:2433-2449. [PMID: 30091661 PMCID: PMC6233056 DOI: 10.1091/mbc.e18-06-0347] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Disruption to the contractility of cells, including smooth muscle cells of the cardiovascular system and myoepithelial cells of the glandular epithelium, contributes to the pathophysiology of contractile tissue diseases, including asthma, hypertension, and primary Sjögren's syndrome. Cell contractility is determined by myosin activity and actomyosin network organization and is mediated by hundreds of protein-protein interactions, many directly involving actin. Here we use a candidate RNA interference screen of more than 100 Caenorhabditis elegans genes with predicted actin-binding and regulatory domains to identify genes that contribute to the contractility of the somatic gonad. We identify the spectrin cytoskeleton composed of SPC-1/α-spectrin, UNC-70/β-spectrin, and SMA-1/β heavy-spectrin as required for contractility and actin organization in the myoepithelial cells of the C. elegans spermatheca. We use imaging of fixed and live animals as well as tissue- and developmental-stage-specific disruption of the spectrin cytoskeleton to show that spectrin regulates the production of prominent central actin bundles and is required for maintenance of central actin bundles throughout successive rounds of stretch and contraction. We conclude that the spectrin cytoskeleton contributes to spermathecal contractility by promoting maintenance of the robust actomyosin bundles that drive contraction.
Collapse
Affiliation(s)
| | - Erin J Cram
- Department of Biology, Northeastern University, Boston, MA 02115
| |
Collapse
|
8
|
Kelley CA, Wirshing ACE, Zaidel-Bar R, Cram EJ. The myosin light-chain kinase MLCK-1 relocalizes during Caenorhabditis elegans ovulation to promote actomyosin bundle assembly and drive contraction. Mol Biol Cell 2018; 29:1975-1991. [PMID: 30088798 PMCID: PMC6232974 DOI: 10.1091/mbc.e18-01-0056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
We identify the Caenorhabditis elegans myosin light-chain kinase, MLCK-1, required for contraction of spermathecae. During contraction, MLCK-1 moves from the apical cell boundaries to the basal actomyosin bundles, where it stabilizes myosin downstream of calcium signaling. MLCK and ROCK act in distinct subsets of cells to coordinate the timing of contraction.
Collapse
Affiliation(s)
| | | | - Ronen Zaidel-Bar
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Erin J Cram
- Department of Biology, Northeastern University, Boston, MA 02115
| |
Collapse
|
9
|
Huang C, Zhang Z, Wang L, Liu J, Gong X, Zhang C. ML-7 attenuates airway inflammation and remodeling via inhibiting the secretion of Th2 cytokines in mice model of asthma. Mol Med Rep 2018; 17:6293-6300. [PMID: 29512725 PMCID: PMC5928606 DOI: 10.3892/mmr.2018.8683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/01/2017] [Indexed: 01/05/2023] Open
Abstract
Previous studies have indicated that smooth muscle myosin light chain kinase (MLCK) has a prominent role in the regulation of smooth muscle contraction, which tends to be upregulated in asthma. In recent years, numerous studies have reported that MLCK is intimately connected with the immunoregulatory mechanism of T cells. The imbalance of T helper type 1 cells (Th1)/Th2 constitutes the immune-associated pathological basis of chronic asthma. Th2-associated cytokines, including interleukin-4, −5, −13, −25 and −33, are involved in airway inflammation, hyperresponsiveness and remodeling, which leads to a progressive decline in lung function. The purpose of the present study was to verify whether inhibition of bronchial MLCK attenuated the expression Th2-associated cytokines in asthmatic mice, including the above-mentioned ones. Female BALB/c mice were used to establish an ovalbumin (OVA)-induced model of asthma, of which one group was treated with the MLCK inhibitor (5-iodonaphthalene-1-sulfonyl) homopiperazine (ML-7). The inhibitor of MLCK, ML-7 attenuated airway inflammation and remodeling by reducing inflammatory cell infiltration and the secretion of Th2 cytokines in mice model of asthma, which may represent a promising therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Chuanjun Huang
- Department of Respiratory Diseases, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, P.R. China
| | - Zewen Zhang
- Department of Medical Imaging and Nuclear Medicine, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Liuxin Wang
- Department of Respiratory Medicine, The First People's Hospital of Jining, Jining, Shandong 272011, P.R. China
| | - Ju Liu
- Department of Medical Research Center, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Xiaodan Gong
- Department of Respiratory Diseases, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Caiqing Zhang
- Department of Respiratory Diseases, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
10
|
Wang T, Brown ME, Kelly GT, Camp SM, Mascarenhas JB, Sun X, Dudek SM, Garcia JGN. Myosin light chain kinase ( MYLK) coding polymorphisms modulate human lung endothelial cell barrier responses via altered tyrosine phosphorylation, spatial localization, and lamellipodial protrusions. Pulm Circ 2018; 8:2045894018764171. [PMID: 29480069 PMCID: PMC5846938 DOI: 10.1177/2045894018764171] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a potent bioactive endogenous lipid that signals a rearrangement of the actin cytoskeleton via the regulation of non-muscle myosin light chain kinase isoform (nmMLCK). S1P induces critical nmMLCK Y464 and Y471 phosphorylation resulting in translocation of nmMLCK to the periphery where spatially-directed increases in myosin light chain (MLC) phosphorylation and tension result in lamellipodia protrusion, increased cell-cell adhesion, and enhanced vascular barrier integrity. MYLK, the gene encoding nmMLCK, is a known candidate gene in lung inflammatory diseases, with coding genetic variants (Pro21His, Ser147Pro, Val261Ala) that confer risk for inflammatory lung injury and influence disease severity. The functional mechanisms by which these MYLK coding single nucleotide polymorphisms (SNPs) affect biologic processes to increase disease risk and severity remain elusive. In the current study, we utilized quantifiable cell immunofluorescence assays to determine the influence of MYLK coding SNPs on S1P-mediated nmMLCK phosphorylation and translocation to the human lung endothelial cell (EC) periphery . These disease-associated MYLK variants result in reduced levels of S1P-induced Y464 phosphorylation, a key site for nmMLCK enzymatic regulation and activation. Reduced Y464 phosphorylation resulted in attenuated nmMLCK protein translocation to the cell periphery. We further conducted EC kymographic assays which confirmed that lamellipodial protrusion in response to S1P challenge was retarded by expression of a MYLK transgene harboring the three MYLK coding SNPs. These data suggest that ARDS/severe asthma-associated MYLK SNPs functionally influence vascular barrier-regulatory cytoskeletal responses via direct alterations in the levels of nmMLCK tyrosine phosphorylation, spatial localization, and lamellipodial protrusions.
Collapse
Affiliation(s)
- Ting Wang
- 1 Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Mary E Brown
- 2 Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Gabriel T Kelly
- 1 Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Sara M Camp
- 1 Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Joseph B Mascarenhas
- 1 Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Xiaoguang Sun
- 1 Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Steven M Dudek
- 2 Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Joe G N Garcia
- 1 Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| |
Collapse
|
11
|
Talwar A, Garcia JGN, Tsai H, Moreno M, Lahm T, Zamanian RT, Machado R, Kawut SM, Selej M, Mathai S, D'Anna LH, Sahni S, Rodriquez EJ, Channick R, Fagan K, Gray M, Armstrong J, Rodriguez Lopez J, de Jesus Perez V. Health Disparities in Patients with Pulmonary Arterial Hypertension: A Blueprint for Action. An Official American Thoracic Society Statement. Am J Respir Crit Care Med 2017; 196:e32-e47. [PMID: 29028375 DOI: 10.1164/rccm.201709-1821st] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Health disparities have a major impact in the quality of life and clinical care received by minorities in the United States. Pulmonary arterial hypertension (PAH) is a rare cardiopulmonary disorder that affects children and adults and that, if untreated, results in premature death. The impact of health disparities in the diagnosis, treatment, and clinical outcome of patients with PAH has not been systematically investigated. OBJECTIVES The specific goals of this research statement were to conduct a critical review of the literature concerning health disparities in PAH, identify major research gaps and prioritize direction for future research. METHODS Literature searches from multiple reference databases were performed using medical subject headings and text words for pulmonary hypertension and health disparities. Members of the committee discussed the evidence and provided recommendations for future research. RESULTS Few studies were found discussing the impact of health disparities in PAH. Using recent research statements focused on health disparities, the group identified six major study topics that would help address the contribution of health disparities to PAH. Representative studies in each topic were discussed and specific recommendations were made by the group concerning the most urgent questions to address in future research studies. CONCLUSIONS At present, there are few studies that address health disparities in PAH. Given the potential adverse impact of health disparities, we recommend that research efforts be undertaken to address the topics discussed in the document. Awareness of health disparities will likely improve advocacy efforts, public health policy and the quality of care of vulnerable populations with PAH.
Collapse
|
12
|
Mascarenhas JB, Tchourbanov AY, Fan H, Danilov SM, Wang T, Garcia JGN. Mechanical Stress and Single Nucleotide Variants Regulate Alternative Splicing of the MYLK Gene. Am J Respir Cell Mol Biol 2017; 56:29-37. [PMID: 27529643 DOI: 10.1165/rcmb.2016-0053oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The nonmuscle (nm) myosin light-chain kinase isoform (MLCK), encoded by the MYLK gene, is a vital participant in regulating vascular barrier responses to mechanical and inflammatory stimuli. We determined that MYLK is alternatively spliced, yielding functionally distinct nmMLCK splice variants including nmMLCK2, a splice variant highly expressed in vascular endothelial cells (EC) and associated with reduced EC barrier integrity. We demonstrated previously that the nmMLCK2 variant lacks exon 11, which encodes a key regulatory region containing two differentially phosphorylated tyrosine residues (Y464 and Y471) that influence vascular barrier function during inflammation. In this study, we used minigene constructs and RT-PCR to interrogate biophysical factors (mechanical stress) and genetic variants (MYLK single-nucleotide polymorphisms [SNPs]) that are potentially involved in regulating MYLK alternative splicing and nmMLCK2 generation. Human lung EC exposed to pathologic mechanical stress (18% cyclic stretch) produced increased nmMLCK2 expression relative to levels of nmMLCK1 with alternative splicing significantly influenced by MYLK SNPs rs77323602 and rs147245669. In silico analyses predicted that these variants would alter exon 11 donor and acceptor sites for alternative splicing, computational predictions that were confirmed by minigene studies. The introduction of rs77323602 favored wild-type nmMLCK expression, whereas rs147245669 favored alternative splicing and deletion of exon 11, yielding increased nmMLCK2 expression. Finally, lymphoblastoid cell lines selectively harboring these MYLK SNPs (rs77323602 and rs147245669) directly validated SNP-specific effects on MYLK alternative splicing and nmMLCK2 generation. Together, these studies demonstrate that mechanical stress and MYLK SNPs regulate MYLK alternative splicing and generation of a splice variant, nmMLCK2, that contributes to the severity of inflammatory injury.
Collapse
Affiliation(s)
| | - Alex Y Tchourbanov
- 2 Arizona Research Laboratory, University of Arizona, Tucson, Arizona; and
| | - Hanli Fan
- 3 Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois
| | - Sergei M Danilov
- 1 Department of Medicine, and.,3 Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois
| | | | | |
Collapse
|
13
|
Wang T, Gross C, Desai AA, Zemskov E, Wu X, Garcia AN, Jacobson JR, Yuan JXJ, Garcia JGN, Black SM. Endothelial cell signaling and ventilator-induced lung injury: molecular mechanisms, genomic analyses, and therapeutic targets. Am J Physiol Lung Cell Mol Physiol 2016; 312:L452-L476. [PMID: 27979857 DOI: 10.1152/ajplung.00231.2016] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/08/2016] [Accepted: 12/11/2016] [Indexed: 12/13/2022] Open
Abstract
Mechanical ventilation is a life-saving intervention in critically ill patients with respiratory failure due to acute respiratory distress syndrome (ARDS). Paradoxically, mechanical ventilation also creates excessive mechanical stress that directly augments lung injury, a syndrome known as ventilator-induced lung injury (VILI). The pathobiology of VILI and ARDS shares many inflammatory features including increases in lung vascular permeability due to loss of endothelial cell barrier integrity resulting in alveolar flooding. While there have been advances in the understanding of certain elements of VILI and ARDS pathobiology, such as defining the importance of lung inflammatory leukocyte infiltration and highly induced cytokine expression, a deep understanding of the initiating and regulatory pathways involved in these inflammatory responses remains poorly understood. Prevailing evidence indicates that loss of endothelial barrier function plays a primary role in the development of VILI and ARDS. Thus this review will focus on the latest knowledge related to 1) the key role of the endothelium in the pathogenesis of VILI; 2) the transcription factors that relay the effects of excessive mechanical stress in the endothelium; 3) the mechanical stress-induced posttranslational modifications that influence key signaling pathways involved in VILI responses in the endothelium; 4) the genetic and epigenetic regulation of key target genes in the endothelium that are involved in VILI responses; and 5) the need for novel therapeutic strategies for VILI that can preserve endothelial barrier function.
Collapse
Affiliation(s)
- Ting Wang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Christine Gross
- Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Ankit A Desai
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Evgeny Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Alexander N Garcia
- Department of Pharmacology University of Illinois at Chicago, Chicago, Illinois; and
| | - Jeffrey R Jacobson
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Jason X-J Yuan
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona;
| |
Collapse
|
14
|
Alcala DB, Haldeman BD, Brizendine RK, Krenc AK, Baker JE, Rock RS, Cremo CR. Myosin light chain kinase steady-state kinetics: comparison of smooth muscle myosin II and nonmuscle myosin IIB as substrates. Cell Biochem Funct 2016; 34:469-474. [PMID: 27528075 DOI: 10.1002/cbf.3209] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 07/07/2016] [Accepted: 07/11/2016] [Indexed: 01/31/2023]
Abstract
Myosin light chain kinase (MLCK) phosphorylates S19 of the myosin regulatory light chain (RLC), which is required to activate myosin's ATPase activity and contraction. Smooth muscles are known to display plasticity in response to factors such as inflammation, developmental stage, or stress, which lead to differential expression of nonmuscle and smooth muscle isoforms. Here, we compare steady-state kinetics parameters for phosphorylation of different MLCK substrates: (1) nonmuscle RLC, (2) smooth muscle RLC, and heavy meromyosin subfragments of (3) nonmuscle myosin IIB, and (4) smooth muscle myosin II. We show that MLCK has a ~2-fold higher kcat for both smooth muscle myosin II substrates compared with nonmuscle myosin IIB substrates, whereas Km values were very similar. Myosin light chain kinase has a 1.6-fold and 1.5-fold higher specificity (kcat /Km ) for smooth versus nonmuscle-free RLC and heavy meromyosin, respectively, suggesting that differences in specificity are dictated by RLC sequences. Of the 10 non-identical RLC residues, we ruled out 7 as possible underlying causes of different MLCK kinetics. The remaining 3 residues were found to be surface exposed in the N-terminal half of the RLC, consistent with their importance in substrate recognition. These data are consistent with prior deletion/chimera studies and significantly add to understanding of MLCK myosin interactions. SIGNIFICANCE OF THE STUDY Phosphorylation of nonmuscle and smooth muscle myosin by myosin light chain kinase (MLCK) is required for activation of myosin's ATPase activity. In smooth muscles, nonmuscle myosin coexists with smooth muscle myosin, but the two myosins have very different chemo-mechanical properties relating to their ability to maintain force. Differences in specificity of MLCK for different myosin isoforms had not been previously investigated. We show that the MLCK prefers smooth muscle myosin by a significant factor. These data suggest that nonmuscle myosin is phosphorylated more slowly than smooth muscle myosin during a contraction cycle.
Collapse
Affiliation(s)
- Diego B Alcala
- Department of Pharmacology, University of Nevada Reno School of Medicine, Reno, Nevada, USA
| | - Brian D Haldeman
- Department of Pharmacology, University of Nevada Reno School of Medicine, Reno, Nevada, USA
| | - Richard K Brizendine
- Department of Pharmacology, University of Nevada Reno School of Medicine, Reno, Nevada, USA
| | - Agata K Krenc
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA
| | - Josh E Baker
- Department of Pharmacology, University of Nevada Reno School of Medicine, Reno, Nevada, USA
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA
| | - Christine R Cremo
- Department of Pharmacology, University of Nevada Reno School of Medicine, Reno, Nevada, USA.
| |
Collapse
|
15
|
Wang T, Mathew B, Wu X, Shimizu Y, Rizzo AN, Dudek SM, Weichselbaum RR, Jacobson JR, Hecker L, Garcia JGN. Nonmuscle myosin light chain kinase activity modulates radiation-induced lung injury. Pulm Circ 2016; 6:234-9. [PMID: 27252850 DOI: 10.1086/686491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Radiotherapy as a primary treatment for thoracic malignancies induces deleterious effects, such as acute or subacute radiation-induced lung injury (RILI). Although the molecular etiology of RILI is controversial and likely multifactorial, a potentially important cellular target is the lung endothelial cytoskeleton that regulates paracellular gap formation and the influx of macromolecules and fluid to the alveolar space. Here we investigate the central role of a key endothelial cytoskeletal regulatory protein, the nonmuscle isoform of myosin light chain kinase (nmMLCK), in an established murine RILI model. Our results indicate that thoracic irradiation significantly augmented nmMLCK protein expression and enzymatic activity in murine lungs. Furthermore, genetically engineered mice harboring a deletion of the nmMLCK gene (nmMLCK(-/-) mice) exhibited protection from RILI, as assessed by attenuated vascular leakage and leukocyte infiltration. In addition, irradiated wild-type mice treated with two distinct MLCK enzymatic inhibitors, ML-7 and PIK (peptide inhibitor of kinase), also demonstrated attenuated RILI. Taken together, these data suggests a key role for nmMLCK in vascular barrier regulation in RILI and warrants further examination of RILI strategies that target nmMLCK.
Collapse
Affiliation(s)
- Ting Wang
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA; These authors contributed equally as first authors
| | - Biji Mathew
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA; These authors contributed equally as first authors
| | - Xiaomin Wu
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA
| | - Yuka Shimizu
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA
| | - Alicia N Rizzo
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Steven M Dudek
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Jeffrey R Jacobson
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Louise Hecker
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA; Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona, USA
| | - Joe G N Garcia
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
16
|
Shimizu Y, Camp SM, Sun X, Zhou T, Wang T, Garcia JGN. Sp1-mediated nonmuscle myosin light chain kinase expression and enhanced activity in vascular endothelial growth factor-induced vascular permeability. Pulm Circ 2015; 5:707-15. [PMID: 26697178 DOI: 10.1086/684124] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Despite the important role played by the nonmuscle isoform of myosin light chain kinase (nmMLCK) in vascular barrier regulation and the implication of both nmMLCK and vascular endothelial growth factor (VEGF) in the pathogenesis of acute respiratory distress syndrome (ARDS), the role played by nmMLCK in VEGF-induced vascular permeability is poorly understood. In this study, the role played by nmMLCK in VEGF-induced vascular hyperpermeability was investigated. Human lung endothelial cell barrier integrity in response to VEGF is examined in both the absence and the presence of nmMLCK small interfering RNAs. Levels of nmMLCK messenger RNA (mRNA), protein, and promoter activity expression were monitored after VEGF stimulation in lung endothelial cells. nmMYLK promoter activity was assessed using nmMYLK promoter luciferase reporter constructs with a series of nested deletions. nmMYLK transcriptional regulation was further characterized by examination of a key transcriptional factor. nmMLCK plays an important role in VEGF-induced permeability. We found that activation of the VEGF signaling pathway in lung endothelial cells increases MYLK gene product at both mRNA and protein levels. Increased nmMLCK mRNA and protein expression is a result of increased nmMYLK promoter activity, regulated in part by binding of the Sp1 transcription factor on triggering by the VEGF signaling pathway. Taken together, these findings suggest that MYLK is an important ARDS candidate gene and a therapeutic target that is highly influenced by excessive VEGF concentrations in the inflamed lung.
Collapse
Affiliation(s)
- Yuka Shimizu
- Department of Medicine and University of Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA
| | - Sara M Camp
- Department of Medicine and University of Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA
| | - Xiaoguang Sun
- Department of Medicine and University of Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA
| | - Tong Zhou
- Department of Medicine and University of Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA
| | - Ting Wang
- Department of Medicine and University of Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA
| | - Joe G N Garcia
- Department of Medicine and University of Arizona Respiratory Center, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
17
|
Acosta-Herrera M, Pino-Yanes M, Ma SF, Barreto-Luis A, Corrales A, Cumplido J, Pérez-Rodríguez E, Campo P, Eng C, García-Robaina JC, Quintela I, Villar J, Blanca M, Carracedo Á, Carrillo T, Garcia JGN, Torgerson DG, Burchard EG, Flores C. Fine mapping of the myosin light chain kinase (MYLK) gene replicates the association with asthma in populations of Spanish descent. J Allergy Clin Immunol 2015; 136:1116-8.e9. [PMID: 26025125 PMCID: PMC4699578 DOI: 10.1016/j.jaci.2015.04.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 04/01/2015] [Accepted: 04/06/2015] [Indexed: 11/18/2022]
Affiliation(s)
- Marialbert Acosta-Herrera
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Research Unit, Hospital Universitario N.S. de Candelaria, Tenerife, Spain; Multidisciplinary Organ Dysfunction Evaluation Research Network (MODERN), Research Unit, Hospital Universitario Dr. Negrin, Gran Canaria, Spain
| | - Maria Pino-Yanes
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Research Unit, Hospital Universitario N.S. de Candelaria, Tenerife, Spain; Department of Medicine, University of California, San Francisco, Calif
| | - Shwu-Fan Ma
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Ill
| | - Amalia Barreto-Luis
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Research Unit, Hospital Universitario N.S. de Candelaria, Tenerife, Spain
| | - Almudena Corrales
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Research Unit, Hospital Universitario N.S. de Candelaria, Tenerife, Spain
| | - José Cumplido
- Allergy Unit, Hospital Universitario Dr. Negrin, Gran Canaria, Spain
| | | | - Paloma Campo
- U.G.C. Allergy, Regional University Hospital of Málaga-IBIMA, Málaga, Spain
| | - Celeste Eng
- Department of Medicine, University of California, San Francisco, Calif
| | | | - Inés Quintela
- Grupo de Medicina Xenómica, CEGEN-ISCIII-Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Jesús Villar
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Multidisciplinary Organ Dysfunction Evaluation Research Network (MODERN), Research Unit, Hospital Universitario Dr. Negrin, Gran Canaria, Spain
| | - Miguel Blanca
- U.G.C. Allergy, Regional University Hospital of Málaga-IBIMA, Málaga, Spain
| | - Ángel Carracedo
- Grupo de Medicina Xenómica, CIBERER-Universidade de Santiago de Compostela-Fundación Galega de Medicina Xenómica (SERGAS), Santiago de Compostela, Spain
| | - Teresa Carrillo
- Allergy Unit, Hospital Universitario Dr. Negrin, Gran Canaria, Spain
| | - Joe G N Garcia
- Arizona Health Sciences Center, University of Arizona, Tucson, Ariz
| | - Dara G Torgerson
- Department of Medicine, University of California, San Francisco, Calif
| | - Esteban G Burchard
- Department of Medicine, University of California, San Francisco, Calif; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, Calif
| | - Carlos Flores
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Research Unit, Hospital Universitario N.S. de Candelaria, Tenerife, Spain; Applied Genomics Group (G2A), Genetics Laboratory, Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
18
|
Shen K, Ramirez B, Mapes B, Shen GR, Gokhale V, Brown ME, Santarsiero B, Ishii Y, Dudek SM, Wang T, Garcia JGN. Structure-Function Analysis of the Non-Muscle Myosin Light Chain Kinase (nmMLCK) Isoform by NMR Spectroscopy and Molecular Modeling: Influence of MYLK Variants. PLoS One 2015; 10:e0130515. [PMID: 26111161 PMCID: PMC4482139 DOI: 10.1371/journal.pone.0130515] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/22/2015] [Indexed: 01/29/2023] Open
Abstract
The MYLK gene encodes the multifunctional enzyme, myosin light chain kinase (MLCK), involved in isoform-specific non-muscle and smooth muscle contraction and regulation of vascular permeability during inflammation. Three MYLK SNPs (P21H, S147P, V261A) alter the N-terminal amino acid sequence of the non-muscle isoform of MLCK (nmMLCK) and are highly associated with susceptibility to acute lung injury (ALI) and asthma, especially in individuals of African descent. To understand the functional effects of SNP associations, we examined the N-terminal segments of nmMLCK by 1H-15N heteronuclear single quantum correlation (HSQC) spectroscopy, a 2-D NMR technique, and by in silico molecular modeling. Both NMR analysis and molecular modeling indicated SNP localization to loops that connect the immunoglobulin-like domains of nmMLCK, consistent with minimal structural changes evoked by these SNPs. Molecular modeling analysis identified protein-protein interaction motifs adversely affected by these MYLK SNPs including binding by the scaffold protein 14-3-3, results confirmed by immunoprecipitation and western blot studies. These structure-function studies suggest novel mechanisms for nmMLCK regulation, which may confirm MYLK as a candidate gene in inflammatory lung disease and advance knowledge of the genetic underpinning of lung-related health disparities.
Collapse
Affiliation(s)
- Kui Shen
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Benjamin Ramirez
- Center for Structural Biology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Brandon Mapes
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Grace R. Shen
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Vijay Gokhale
- College of Pharmacy and BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Mary E. Brown
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Bernard Santarsiero
- Center for Structural Biology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Yoshitaka Ishii
- Center for Structural Biology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Steven M. Dudek
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ting Wang
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Arizona Respiratory Center and Department of Medicine, University of Arizona, Tucson, Arizona, United States of America
| | - Joe G. N. Garcia
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Arizona Respiratory Center and Department of Medicine, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
19
|
Zhou T, Wang T, Garcia JGN. A nonmuscle myosin light chain kinase-dependent gene signature in peripheral blood mononuclear cells is linked to human asthma severity and exacerbation status. Pulm Circ 2015; 5:335-8. [PMID: 26064459 DOI: 10.1086/680357] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/13/2014] [Indexed: 12/12/2022] Open
Abstract
Asthma is increasingly recognized as a heterogeneous disease influenced by complex genetic and environmental contributions. Myosin light chain kinase (MLCK; gene symbol, MYLK), especially the nonmuscle isoform nmMLCK, is a cytoskeleton protein known to be related to human asthma susceptibility and severity, findings confirmed in preclinical models of asthmatic inflammation. In this study, we define the central capacity for a nmMLCK-influenced gene signature in human peripheral blood mononuclear cells to predict human asthma severity and exacerbation status. We refined this signature from a list of nmMLCK-influenced genes identified in lung tissues of nmMLCK knockout mice exposed to inflammatory stimuli (ventilator-induced lung injury), with subsequent identification of nmMLCK-influenced genes in a list of human asthma severity-related genes expressed in blood. The enriched nmMLCK-influenced gene signature successfully predicted human asthma severity and exacerbation status in both discovery and validation human asthma cohorts. These findings validate the central role played by nmMLCK in asthma susceptibility, severity, and exacerbation and further provide novel gene signatures as effective asthma biomarkers for severity, exacerbation, and prognosis.
Collapse
Affiliation(s)
- Tong Zhou
- Arizona Respiratory Center and Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Ting Wang
- TW and JGNG contributed equally as senior authors
| | | |
Collapse
|
20
|
A MYLK variant regulates asthmatic inflammation via alterations in mRNA secondary structure. Eur J Hum Genet 2014; 23:874-6. [PMID: 25271083 PMCID: PMC4795064 DOI: 10.1038/ejhg.2014.201] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 08/20/2014] [Accepted: 08/26/2014] [Indexed: 11/30/2022] Open
Abstract
Myosin light-chain kinase (MYLK) is a gene known to be significantly associated with severe asthma in African Americans. Here we further examine the molecular function of a single-nucleotide polymorphism (SNP), located in the non-muscle myosin light-chain kinase isoform (nmMLCK), in asthma susceptibility and pathobiology. We identified nmMLCK variant (reference SNP: rs9840993, NM_053025: 721C>T, c.439C>T) with a distinct mRNA secondary structure from the other variants. The nmMLCK variant (721C) secondary structure exhibits increased stability with an elongated half-life in the human endothelial cell, and greater efficiency in protein translation initiation owing to an increased accessibility to translation start site. Finally, nmMLCK expression of 721C- and 721T-containing MYLK transgenes were compared in nmMLCK−/− mice and confirmed deleterious effects of nmMLCK expression on asthmatic indices and implicated the augmented influence of MYLK 721C>T (c.439C>T) SNP on asthma severity. The confirmation of the novel mechanism of the regulation of asthmatic inflammation by a MYLK advances knowledge of the genetic basis for asthma disparities, and further suggests the potential of nmMLCK as a therapeutic target. Our study suggests that in addition to altering protein structure and function, non-synonymous SNPs may also lead to phenotypic disparity by altering protein expression.
Collapse
|
21
|
Wang T, Moreno-Vinasco L, Ma SF, Zhou T, Shimizu Y, Sammani S, Epshtein Y, Watterson DM, Dudek SM, Garcia JGN. Nonmuscle myosin light chain kinase regulates murine asthmatic inflammation. Am J Respir Cell Mol Biol 2014; 50:1129-35. [PMID: 24428690 PMCID: PMC4068916 DOI: 10.1165/rcmb.2013-0434oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Myosin light chain kinase (MLCK; gene code, MYLK) is a multifunctional enzyme involved in isoform-specific nonmuscle (nm) and smooth muscle contraction, inflammation, and vascular permeability, processes directly relevant to asthma pathobiology. In this report, we highlight the contribution of the nm isoform (nmMLCK) to asthma susceptibility and severity, supported by studies in two lines of transgenic mice with knocking out nmMLCK or selectively overexpressing nmMLCK in endothelium. These mice were sensitized to exhibit ovalbumin-mediated allergic inflammation. Genetically engineered mice with targeted nmMLCK deletion (nmMLCK(-/-)) exhibited significant reductions in lung inflammation and airway hyperresponsiveness. Conversely, mice with overexpressed nmMLCK in endothelium (nmMLCK(ec/ec)) exhibited elevated susceptibility and severity in asthmatic inflammation. In addition, reduction of nmMLCK expression in pulmonary endothelium by small interfering RNA results in reduced asthmatic inflammation in wild-type mice. These pathophysiological assessments demonstrate the positive contribution of nmMLCK to asthmatic inflammation, and a clear correlation of the level of nmMLCK with the degree of experimental allergic inflammation. This study confirms MYLK as an asthma candidate gene, and verifies nmMLCK as a novel molecular target in asthmatic pathobiology.
Collapse
Affiliation(s)
- Ting Wang
- 1 Arizona Respiratory Center and Department of Medicine, University of Arizona, Tucson, Arizona
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Betapudi V. Life without double-headed non-muscle myosin II motor proteins. Front Chem 2014; 2:45. [PMID: 25072053 PMCID: PMC4083560 DOI: 10.3389/fchem.2014.00045] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 06/19/2014] [Indexed: 11/20/2022] Open
Abstract
Non-muscle myosin II motor proteins (myosin IIA, myosin IIB, and myosin IIC) belong to a class of molecular motor proteins that are known to transduce cellular free-energy into biological work more efficiently than man-made combustion engines. Nature has given a single myosin II motor protein for lower eukaryotes and multiple for mammals but none for plants in order to provide impetus for their life. These specialized nanomachines drive cellular activities necessary for embryogenesis, organogenesis, and immunity. However, these multifunctional myosin II motor proteins are believed to go awry due to unknown reasons and contribute for the onset and progression of many autosomal-dominant disorders, cataract, deafness, infertility, cancer, kidney, neuronal, and inflammatory diseases. Many pathogens like HIV, Dengue, hepatitis C, and Lymphoma viruses as well as Salmonella and Mycobacteria are now known to take hostage of these dedicated myosin II motor proteins for their efficient pathogenesis. Even after four decades since their discovery, we still have a limited knowledge of how these motor proteins drive cell migration and cytokinesis. We need to enrich our current knowledge on these fundamental cellular processes and develop novel therapeutic strategies to fix mutated myosin II motor proteins in pathological conditions. This is the time to think how to relieve the hijacked myosins from pathogens in order to provide a renewed impetus for patients' life. Understanding how to steer these molecular motors in proliferating and differentiating stem cells will improve stem cell based-therapeutics development. Given the plethora of cellular activities non-muscle myosin motor proteins are involved in, their importance is apparent for human life.
Collapse
Affiliation(s)
- Venkaiah Betapudi
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic Cleveland, OH, USA ; Department of Physiology and Biophysics, Case Western Reserve University Cleveland, OH, USA
| |
Collapse
|
23
|
Variants of CEP68 gene are associated with acute urticaria/angioedema induced by multiple non-steroidal anti-inflammatory drugs. PLoS One 2014; 9:e90966. [PMID: 24618698 PMCID: PMC3949706 DOI: 10.1371/journal.pone.0090966] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 02/06/2014] [Indexed: 01/18/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are the most consumed drugs worldwide because of their efficacy and utility in the treatment of pain and inflammatory diseases. However, they are also responsible for an important number of adverse effects including hypersensitivity reactions. The most important group of these reactions is triggered by non-immunological, pharmacological mechanisms catalogued under the denomination of cross-intolerance (CRI), with acute urticaria/angioedema induced by multiple NSAIDs (MNSAID-UA) the most frequently associated clinical entity. A recent genome-wide association study identified the gene encoding the centrosomal protein of 68 KDa (CEP68) as the major locus associated with aspirin intolerance susceptibility in asthmatics. In this study, we aimed to assess the role of this locus in susceptibility to CRI to NSAIDs by examining 53 common gene variants in a total of 635 patients that were classified as MNSAID-UA (n = 399), airway exacerbations (n = 110) or blended pattern (n = 126), and 425 controls. We found in the MNSAID-UA group a number of variants (17) associated (lowest p-value = 1.13×10−6), including the non-synonymous Gly74Ser variant (rs7572857) previously associated with aspirin intolerance susceptibility in asthmatics. Although not being significant in the context of multiple testing, eight of these variants were also associated with exacerbated respiratory disease or blended reactions. Our results suggest that CEP68 gene variants may play an important role in MNSAID-UA susceptibility and, despite the different regulatory mechanisms involved depending on the specific affected organ, in the development of hypersensitivity reactions to NSAIDs.
Collapse
|
24
|
Park S, Park MS, Jung KH, Song J, Kim YA, Cho HJ, Min BI, Bae H. Treatment with pyranopyran-1, 8-dione attenuates airway responses in cockroach allergen sensitized asthma in mice. PLoS One 2014; 9:e87558. [PMID: 24489937 PMCID: PMC3906187 DOI: 10.1371/journal.pone.0087558] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 12/24/2013] [Indexed: 12/25/2022] Open
Abstract
Chronic allergic asthma is characterized by Th2-typed inflammation, and contributes to airway remodeling and the deterioration of lung function. Viticis Fructus (VF) has long been used in China and Korea as a traditional herbal remedy for treating various inflammatory diseases. Previously, we have isolated a novel phytochemical, pyranopyran-1, 8-dione (PPY), from VF. This study was conducted to evaluate the ability of PPY to prevent airway inflammation and to attenuate airway responses in a cockroach allergen-induced asthma model in mice. The mice sensitized to and challenged with cockroach allergen were treated with oral administration of PPY. The infiltration of total cells, eosinophils and lymphocytes into the BAL fluid was significantly inhibited in cockroach allergen-induced asthma mice treated with PPY (1, 2, or 10 mg/kg). Th2 cytokines and chemokine, such as IL-4, IL-5, IL-13 and eotaxin in BAL fluid were also reduced to normal levels following treatment with PPY. In addition, the levels of IgE were also markedly suppressed after PPY treatment. Histopathological examination demonstrated that PPY substantially inhibited eosinophil infiltration into the airway, goblet cell hyperplasia and smooth muscle hypertrophy. Taken together, these results demonstrate that PPY possesses a potent efficacy on controlling allergic asthma response such as airway inflammation and remodeling.
Collapse
Affiliation(s)
- Soojin Park
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Min-Sun Park
- Department of East-West Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Kyung-Hwa Jung
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Joohyun Song
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - You Ah Kim
- Natural Products Department, Korea Promotion Institute for Traditional Medical Industry, Kyungbuk, Republic of Korea
| | - Hi Jae Cho
- Natural Products Department, Korea Promotion Institute for Traditional Medical Industry, Kyungbuk, Republic of Korea
| | - Byung-Il Min
- Department of East-West Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyunsu Bae
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
25
|
Wu Q, Chu HW. Role of infections in the induction and development of asthma: genetic and inflammatory drivers. Expert Rev Clin Immunol 2014; 5:97-109. [PMID: 19885377 DOI: 10.1586/1744666x.5.1.97] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Genetic and environmental factors interact to initiate and even maintain the course of asthma. As one of the highly risky environmental factors, infections in predisposed individuals can promote asthma development and exacerbations and/or prolong symptoms. This review will describe our current understanding of the genetic markers of innate immunity in the induction and development of asthma, the diverse roles of infections in modulating allergic inflammation, host susceptibility to infections and subsequent acute exacerbations in an allergic setting, and the therapeutic or preventive implications of existing knowledge. Current challenges and future directions in basic and clinical research of asthma are also discussed.
Collapse
Affiliation(s)
- Qun Wu
- Postdoctoral Research Fellow, Department of Medicine, National Jewish Health, 1400 Jackson Street, Room A635, Denver, CO 80206, USA, Tel.: +1 303 398 1589, ,
| | | |
Collapse
|
26
|
Chen M, Zhang W, Lu X, Hoggatt AM, Gunst SJ, Kassab GS, Tune JD, Herring BP. Regulation of 130-kDa smooth muscle myosin light chain kinase expression by an intronic CArG element. J Biol Chem 2013; 288:34647-57. [PMID: 24151072 DOI: 10.1074/jbc.m113.510362] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The mylk1 gene encodes a 220-kDa nonmuscle myosin light chain kinase (MLCK), a 130-kDa smooth muscle MLCK (smMLCK), as well as the non-catalytic product telokin. Together, these proteins play critical roles in regulating smooth muscle contractility. Changes in their expression are associated with many pathological conditions; thus, it is important to understand the mechanisms regulating expression of mylk1 gene transcripts. Previously, we reported a highly conserved CArG box, which binds serum response factor, in intron 15 of mylk1. Because this CArG element is near the promoter that drives transcription of the 130-kDa smMLCK, we examined its role in regulating expression of this transcript. Results show that deletion of the intronic CArG region from a β-galactosidase reporter gene abolished transgene expression in mice in vivo. Deletion of the CArG region from the endogenous mylk1 gene, specifically in smooth muscle cells, decreased expression of the 130-kDa smMLCK by 40% without affecting expression of the 220-kDa MLCK or telokin. This reduction in 130-kDa smMLCK expression resulted in decreased phosphorylation of myosin light chains, attenuated smooth muscle contractility, and a 24% decrease in small intestine length that was associated with a significant reduction of Ki67-positive smooth muscle cells. Overall, these data show that the CArG element in intron 15 of the mylk1 gene is necessary for maximal expression of the 130-kDa smMLCK and that the 130-kDa smMLCK isoform is specifically required to regulate smooth muscle contractility and small intestine smooth muscle cell proliferation.
Collapse
Affiliation(s)
- Meng Chen
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Sun X, Ma SF, Wade MS, Acosta-Herrera M, Villar J, Pino-Yanes M, Zhou T, Liu B, Belvitch P, Moitra J, Han YJ, Machado R, Noth I, Natarajan V, Dudek SM, Jacobson JR, Flores C, Garcia JGN. Functional promoter variants in sphingosine 1-phosphate receptor 3 associate with susceptibility to sepsis-associated acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol 2013; 305:L467-77. [PMID: 23911438 DOI: 10.1152/ajplung.00010.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The genetic mechanisms underlying the susceptibility to acute respiratory distress syndrome (ARDS) are poorly understood. We previously demonstrated that sphingosine 1-phosphate (S1P) and the S1P receptor S1PR3 are intimately involved in lung inflammatory responses and vascular barrier regulation. Furthermore, plasma S1PR3 protein levels were shown to serve as a biomarker of severity in critically ill ARDS patients. This study explores the contribution of single nucleotide polymorphisms (SNPs) of the S1PR3 gene to sepsis-associated ARDS. S1PR3 SNPs were identified by sequencing the entire gene and tagging SNPs selected for case-control association analysis in African- and ED samples from Chicago, with independent replication in a European case-control study of Spanish individuals. Electrophoretic mobility shift assays, luciferase activity assays, and protein immunoassays were utilized to assess the functionality of associated SNPs. A total of 80 variants, including 29 novel SNPs, were identified. Because of limited sample size, conclusive findings could not be drawn in African-descent ARDS subjects; however, significant associations were found for two promoter SNPs (rs7022797 -1899T/G; rs11137480 -1785G/C), across two ED samples supporting the association of alleles -1899G and -1785C with decreased risk for sepsis-associated ARDS. In addition, these alleles significantly reduced transcription factor binding to the S1PR3 promoter; reduced S1PR3 promoter activity, a response particularly striking after TNF-α challenge; and were associated with lower plasma S1PR3 protein levels in ARDS patients. These highly functional studies support S1PR3 as a novel ARDS candidate gene and a potential target for individualized therapy.
Collapse
Affiliation(s)
- Xiaoguang Sun
- Institute for Personalize Respiratory Medicine, Univ. of Illinois at Chicago, 3099 COMRB (MC719 909 S. Wolcott Ave., Chicago, IL 60612.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Doeing DC, Solway J. Airway smooth muscle in the pathophysiology and treatment of asthma. J Appl Physiol (1985) 2013; 114:834-43. [PMID: 23305987 DOI: 10.1152/japplphysiol.00950.2012] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Airway smooth muscle (ASM) plays an integral part in the pathophysiology of asthma. It is responsible for acute bronchoconstriction, which is potentiated by constrictor hyperresponsiveness, impaired relaxation and length adaptation. ASM also contributes to airway remodeling and inflammation in asthma. In light of this, ASM is an important target in the treatment of asthma.
Collapse
Affiliation(s)
- Diana C Doeing
- Department of Medicine, University of Chicago, Chicago, IL, USA.
| | | |
Collapse
|
29
|
Accumulating evidence for increased velocity of airway smooth muscle shortening in asthmatic airway hyperresponsiveness. J Allergy (Cairo) 2012; 2012:156909. [PMID: 23319963 PMCID: PMC3540810 DOI: 10.1155/2012/156909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 12/06/2012] [Indexed: 11/18/2022] Open
Abstract
It remains unclear whether airway smooth muscle (ASM) mechanics is altered in asthma. While efforts have originally focussed on contractile force, some evidence points to an increased velocity of shortening. A greater rate of airway renarrowing after a deep inspiration has been reported in asthmatics compared to controls, which could result from a shortening velocity increase. In addition, we have recently shown in rats that increased shortening velocity correlates with increased muscle shortening, without increasing muscle force. Nonetheless, establishing whether or not asthmatic ASM shortens faster than that of normal subjects remains problematic. Endobronchial biopsies provide excellent tissue samples because the patients are well characterized, but the size of the samples allows only cell level experiments. Whole human lungs from transplant programs suffer primarily from poor patient characterization, leading to high variability. ASM from several animal models of asthma has shown increased shortening velocity, but it is unclear whether this is representative of human asthma. Several candidates have been suggested as responsible for increased shortening velocity in asthma, such as alterations in contractile protein expression or changes in the contractile apparatus structure. There is no doubt that more remains to be learned about the role of shortening velocity in asthma.
Collapse
|
30
|
Wan ES, Qiu W, Baccarelli A, Carey VJ, Bacherman H, Rennard SI, Agusti A, Anderson W, Lomas DA, Demeo DL. Cigarette smoking behaviors and time since quitting are associated with differential DNA methylation across the human genome. Hum Mol Genet 2012; 21:3073-82. [PMID: 22492999 DOI: 10.1093/hmg/dds135] [Citation(s) in RCA: 242] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The impact of cigarette smoking can persist for extended periods following smoking cessation and may involve epigenetic reprogramming. Changes in DNA methylation associated with smoking may help to identify molecular pathways that contribute to the latency between exposure and disease onset. Cross-sectional cohort data from subjects in the International COPD Genetics Network (n = 1085) and the Boston Early-Onset COPD study (n = 369) were analyzed as the discovery and replication cohorts, respectively. Genome-wide methylation data on 27 578 CpG sites in 14 475 genes were obtained on DNA from peripheral blood leukocytes using the Illumina HumanMethylation27K Beadchip in both cohorts. We identified 15 sites significantly associated with current smoking, 2 sites associated with cumulative smoke exposure, and, within the subset of former smokers, 3 sites associated with time since quitting cigarettes. Two loci, factor II receptor-like 3 (F2RL3) and G-protein-coupled receptor 15 (GPR15), were significantly associated in all three analyses and were validated by pyrosequencing. These findings (i) identify a novel locus (GPR15) associated with cigarette smoking and (ii) suggest the existence of dynamic, site-specific methylation changes in response to smoking which may contribute to the extended risks associated with cigarette smoking that persist after cessation.
Collapse
Affiliation(s)
- Emily S Wan
- Channing Laboratory and the Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
African ancestry is associated with asthma risk in African Americans. PLoS One 2012; 7:e26807. [PMID: 22235241 PMCID: PMC3250386 DOI: 10.1371/journal.pone.0026807] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 10/04/2011] [Indexed: 11/19/2022] Open
Abstract
Background Asthma is a common complex condition with clear racial and ethnic differences in both prevalence and severity. Asthma consultation rates, mortality, and severe symptoms are greatly increased in African descent populations of developed countries. African ancestry has been associated with asthma, total serum IgE and lower pulmonary function in African-admixed populations. To replicate previous findings, here we aimed to examine whether African ancestry was associated with asthma susceptibility in African Americans. In addition, we examined for the first time whether African ancestry was associated with asthma exacerbations. Methodology/Principal Findings After filtering for self-reported ancestry and genotype data quality, samples from 1,117 self-reported African-American individuals from New York and Baltimore (394 cases, 481 controls), and Chicago (321 cases followed for asthma exacerbations) were analyzed. Genetic ancestry was estimated based on ancestry informative markers (AIMs) selected for being highly divergent among European and West African populations (95 AIMs for New York and Baltimore, and 66 independent AIMs for Chicago). Among case-control samples, the mean African ancestry was significantly higher in asthmatics than in non-asthmatics (82.0±14.0% vs. 77.8±18.1%, mean difference 4.2% [95% confidence interval (CI):2.0–6.4], p<0.0001). This association remained significant after adjusting for potential confounders (odds ratio: 4.55, 95% CI: 1.69–12.29, p = 0.003). African ancestry failed to show an association with asthma exacerbations (p = 0.965) using a model based on longitudinal data of the number of exacerbations followed over 1.5 years. Conclusions/Significance These data replicate previous findings indicating that African ancestry constitutes a risk factor for asthma and suggest that elevated asthma rates in African Americans can be partially attributed to African genetic ancestry.
Collapse
|
32
|
Ma SF, Xie L, Pino-Yanes M, Sammani S, Wade MS, Letsiou E, Siegler J, Wang T, Infusino G, Kittles RA, Flores C, Zhou T, Prabhakar BS, Moreno-Vinasco L, Villar J, Jacobson JR, Dudek SM, Garcia JGN. Type 2 deiodinase and host responses of sepsis and acute lung injury. Am J Respir Cell Mol Biol 2011; 45:1203-11. [PMID: 21685153 PMCID: PMC3262665 DOI: 10.1165/rcmb.2011-0179oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 06/10/2011] [Indexed: 12/21/2022] Open
Abstract
The role of thyroid hormone metabolism in clinical outcomes of the critically ill remains unclear. Using preclinical models of acute lung injury (ALI), we assessed the gene and protein expression of type 2 deiodinase (DIO2), a key driver for synthesis of biologically active triiodothyronine, and addressed potential association of DIO2 genetic variants with ALI in a multiethnic cohort. DIO2 gene and protein expression levels in murine lung were validated by microarrays and immunoblotting. Lung injury was assessed by levels of bronchoalveolar lavage protein and leukocytes. Single-nucleotide polymorphisms were genotyped and ALI susceptibility association assessed. Significant increases in both DIO2 gene and D2 protein expression were observed in lung tissues from murine ALI models (LPS- and ventilator-induced lung injury), with expression directly increasing with the extent of lung injury. Mice with reduced levels of DIO2 expression (by silencing RNA) demonstrated reduced thyroxine levels in plasma and increased lung injury (increased bronchoalveolar lavage protein and leukocytes), suggesting a protective role for DIO2 in ALI. The G (Ala) allele of the Thr92Ala coding single-nucleotide polymorphism (rs225014) was protective in severe sepsis and severe sepsis-associated ALI after adjustments for age, sex, and genetic ancestry in a logistic regression model in European Americans. Our studies indicate that DIO2 is a novel ALI candidate gene, the nonsynonymous Thr92Ala coding variant of which confers ALI protection. Increased DIO2 expression may dampen the ALI inflammatory response, thereby strengthening the premise that thyroid hormone metabolism is intimately linked to the integrated response to inflammatory injury in critically ill patients.
Collapse
Affiliation(s)
- Shwu-Fan Ma
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Lishi Xie
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Maria Pino-Yanes
- Centro de Investigacion Biomedica en Red Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Tenerife, Spain
| | - Saad Sammani
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Michael S. Wade
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Eleftheria Letsiou
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Jessica Siegler
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Ting Wang
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Giovanni Infusino
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Rick A. Kittles
- Section of Hematology and Oncology, Department of Medicine, and
| | - Carlos Flores
- Centro de Investigacion Biomedica en Red Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Tenerife, Spain
| | - Tong Zhou
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Bellur S. Prabhakar
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| | - Liliana Moreno-Vinasco
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Jesus Villar
- Centro de Investigacion Biomedica en Red Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain; and
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Canada
| | - Jeffrey R. Jacobson
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Steven M. Dudek
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Joe G. N. Garcia
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
33
|
An intronic MYLK variant associated with inflammatory lung disease regulates promoter activity of the smooth muscle myosin light chain kinase isoform. J Mol Med (Berl) 2011; 90:299-308. [PMID: 22015949 DOI: 10.1007/s00109-011-0820-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 08/16/2011] [Accepted: 09/08/2011] [Indexed: 10/24/2022]
Abstract
Intronic single-nucleotide polymorphisms (SNPs) are commonly associated with complex diseases but exhibit unknown biologic functionality. Myosin light-chain kinase (MLCK), a central cytoskeletal regulator encoded by MYLK, plays a key pathophysiological role in complex diseases including acute lung injury (ALI) and asthma. We studied the potential regulatory roles of two intronic MYLK SNPs (rs936170 and rs820336) previously associated with ALI and asthma. Due to their genomic location at the junction encoding the non-muscle and smooth muscle MLCK (smMLCK) isoforms, we first identified the transcription start site (TSS) of the smMLCK isoform, and isolated a 2,954-bp DNA fragment upstream of the smMLCK TSS. Serial 5' deletion of the fragment revealed a proximal promoter region exhibiting strong promoter activity with potential inhibitory elements in the distal region. Site-directed mutageneses and luciferase reporter assays showed no effect of the distal promoter SNP rs936170 on smMLCK promoter activity. In contrast, SNP rs820336, located in an enhancer/repressor region downstream of TSS, was identified to regulate smMLCK promoter activity in an allelic-dependent manner. The A allele interrupted the binding site for Forkhead box protein N1 (FOXN1), a transcription factor governing expression of immune response genes. Silencing of FOXN1 expression (siRNA) reduced FOXN1 interaction with cis-regulatory elements in proximity to rs820336 and significantly decreased smMLCK expression. These functional insights into the involvement of intronic MYLK SNPs further strengthen the concept that MYLK contributes to inflammatory disease susceptibility and represents an attractive molecular target in complex inflammatory disorders.
Collapse
|
34
|
Hong F, Haldeman BD, Jackson D, Carter M, Baker JE, Cremo CR. Biochemistry of smooth muscle myosin light chain kinase. Arch Biochem Biophys 2011. [PMID: 21565153 DOI: 10.1016/j.abb.2011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The smooth muscle isoform of myosin light chain kinase (MLCK) is a Ca(2+)-calmodulin-activated kinase that is found in many tissues. It is particularly important for regulating smooth muscle contraction by phosphorylation of myosin. This review summarizes selected aspects of recent biochemical work on MLCK that pertains to its function in smooth muscle. In general, the focus of the review is on new findings, unresolved issues, and areas with the potential for high physiological significance that need further study. The review includes a concise summary of the structure, substrates, and enzyme activity, followed by a discussion of the factors that may limit the effective activity of MLCK in the muscle. The interactions of each of the many domains of MLCK with the proteins of the contractile apparatus, and the multi-domain interactions of MLCK that may control its behaviors in the cell are summarized. Finally, new in vitro approaches to studying the mechanism of phosphorylation of myosin are introduced.
Collapse
Affiliation(s)
- Feng Hong
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, 89557, USA
| | | | | | | | | | | |
Collapse
|
35
|
Biochemistry of smooth muscle myosin light chain kinase. Arch Biochem Biophys 2011; 510:135-46. [PMID: 21565153 DOI: 10.1016/j.abb.2011.04.018] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 04/22/2011] [Accepted: 04/25/2011] [Indexed: 11/23/2022]
Abstract
The smooth muscle isoform of myosin light chain kinase (MLCK) is a Ca(2+)-calmodulin-activated kinase that is found in many tissues. It is particularly important for regulating smooth muscle contraction by phosphorylation of myosin. This review summarizes selected aspects of recent biochemical work on MLCK that pertains to its function in smooth muscle. In general, the focus of the review is on new findings, unresolved issues, and areas with the potential for high physiological significance that need further study. The review includes a concise summary of the structure, substrates, and enzyme activity, followed by a discussion of the factors that may limit the effective activity of MLCK in the muscle. The interactions of each of the many domains of MLCK with the proteins of the contractile apparatus, and the multi-domain interactions of MLCK that may control its behaviors in the cell are summarized. Finally, new in vitro approaches to studying the mechanism of phosphorylation of myosin are introduced.
Collapse
|
36
|
Garcia JGN. Genomic investigations into acute inflammatory lung injury. PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY 2011; 8:167-72. [PMID: 21543796 PMCID: PMC3131835 DOI: 10.1513/pats.201101-002ms] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 03/02/2011] [Indexed: 11/20/2022]
Abstract
Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome, are complex illnesses involving the interplay of both environmental (such as mechanical ventilation) and genetic factors. To understand better the underlying mechanisms of pathogenesis associated with ALI, we recently identified several candidate genes by global expression profiling in preclinical models of ALI and relevant single-nucleotide polymorphisms. We summarize here several strategies successfully used to identify novel ALI candidate genes and detail the validation of variants in these genes as contributing factors to ALI pathobiology, conclusions based on functional analyses, and specific genetic association studies conducted in ALI cohorts. Continued insights into ALI pathogenesis and identification of genetic variants, which confer ALI risk and severity, promise to reveal novel molecular therapeutic targets that can be translated into personalized treatments to reduce the very high, unacceptable mortality of this disorder.
Collapse
|
37
|
Zhou T, Garcia JG, Zhang W. Integrating microRNAs into a system biology approach to acute lung injury. Transl Res 2011; 157:180-90. [PMID: 21420028 PMCID: PMC3073780 DOI: 10.1016/j.trsl.2011.01.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 01/15/2011] [Accepted: 01/18/2011] [Indexed: 01/02/2023]
Abstract
Acute lung injury (ALI), including the ventilator-induced lung injury (VILI) and the more severe acute respiratory distress syndrome (ARDS), are common and complex inflammatory lung diseases potentially affected by various genetic and nongenetic factors. Using the candidate gene approach, genetic variants associated with immune response and inflammatory pathways have been identified and implicated in ALI. Because gene expression is an intermediate phenotype that resides between the DNA sequence variation and the higher level cellular or whole-body phenotypes, the illustration of gene expression regulatory networks potentially could enhance understanding of disease susceptibility and the development of inflammatory lung syndromes. MicroRNAs (miRNAs) have emerged as a novel class of gene regulators that play critical roles in complex diseases including ALI. Comparisons of global miRNA profiles in animal models of ALI and VILI identified several miRNAs (eg, miR-146a and miR-155) previously implicated in immune response and inflammatory pathways. Therefore, via regulation of target genes in these biological processes and pathways, miRNAs potentially contribute to the development of ALI. Although this line of inquiry exists at a nascent stage, miRNAs have the potential to be critical components of a comprehensive model for inflammatory lung disease built by a systems biology approach that integrates genetic, genomic, proteomic, epigenetic as well as environmental stimuli information. Given their particularly recognized role in regulation of immune and inflammatory responses, miRNAs also serve as novel therapeutic targets and biomarkers for ALI/ARDS or VILI, thus facilitating the realization of personalized medicine for individuals with acute inflammatory lung disease.
Collapse
Affiliation(s)
- Tong Zhou
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Joe G.N. Garcia
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wei Zhang
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL 60612, USA
- Institute for Human Genetics, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
38
|
Miller EK, Dumitrescu L, Cupp C, Dorris S, Taylor S, Sparks R, Fawkes D, Frontiero V, Rezendes AM, Marchant C, Edwards KM, Crawford DC. Atopy history and the genomics of wheezing after influenza vaccination in children 6-59 months of age. Vaccine 2011; 29:3431-7. [PMID: 21396408 DOI: 10.1016/j.vaccine.2011.02.071] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 02/13/2011] [Accepted: 02/20/2011] [Indexed: 01/13/2023]
Abstract
BACKGROUND A multinational clinical trial compared the safety and efficacy of intranasal trivalent live attenuated influenza vaccine (LAIV) with intramuscular trivalent inactivated vaccine (TIV) in very young children prior to the 2004-5 influenza season [1]. Wheezing was noted more often in recipients of LAIV and laboratory-confirmed influenza infection was noted more often in recipients of TIV. We sought to determine whether epidemiologic or genetic factors were associated with these outcomes. METHODS Atopy surveys and DNA collections were performed in trial participants at two United States sites, Nashville, TN and Boston, MA. DNA samples were genotyped on Illumina Infinium 610 or 660-Quad. Standard allelic tests of association were performed. RESULTS At the Nashville and Boston sites, a total of 99 children completed the trial, 6 (1 TIV, 5 LAIV) developed medically attended wheezing within 42 days following vaccination, and 8 (5 TIV, 3 LAIV) developed laboratory-confirmed influenza during the season. Eighty-one surveys and 70 DNA samples were collected. Family history of asthma (p=0.001) was associated with wheezing after vaccination. Of 468,458 single nucleotide polymorphisms tested in the genome-wide association study (GWAS), none achieved genome-wide significance for either wheezing after vaccination or laboratory-confirmed influenza infection. CONCLUSIONS Family history of asthma appears to be a risk factor for wheezing after influenza vaccination. Given the limitations of the sample size, our pilot study demonstrated the feasibility of performing a GWAS but was not able to determine genetic polymorphisms associated with wheezing after influenza immunization.
Collapse
Affiliation(s)
- E Kathryn Miller
- Department of Pediatrics, Vanderbilt University Medical Center, CCC-5323 Medical Center North, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yuan JXJ, Garcia JG, West JB, Hales CA, Rich S, Archer SL. Genomics of Acute Lung Injury and Vascular Barrier Dysfunction. TEXTBOOK OF PULMONARY VASCULAR DISEASE 2011. [PMCID: PMC7122529 DOI: 10.1007/978-0-387-87429-6_63] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Acute lung injury (ALI) is a devastating syndrome of diffuse alveolar damage that develops via a variety of local and systemic insults such as sepsis, trauma, pneumonia, and aspiration. It is interestingly to note that only a subset of individuals exposed to potential ALI-inciting insults develop the disorder and the severity of the disease varies from complete resolution to death. In addition, ALI susceptibility and severity are also affected by ethnicity as evidenced by the higher mortality rates observed in African-American ALI patients compared with other ethnic groups in the USA. Moreover, marked differences in strain-specific ALI responses to inflammatory and injurious agents are observed in preclinical animal models. Together, these observations strongly indicate genetic components to be involved in the pathogenesis of ALI. The identification of genes contributing to ALI would potentially provide a better understanding of ALI pathobiology, yield novel biomarkers, identify individuals or populations at risk, and prove useful for the development of novel and individualized therapies. Genome-wide searches in animal models have identified a number of quantitative trait loci that associate with ALI susceptibility. In this chapter, we utilize a systems biology approach combining cellular signaling pathway analysis with population- based association studies to review established and suspected candidate genes that contribute to dysfunction of endothelial cell barrier integrity and ALI susceptibility.
Collapse
Affiliation(s)
- Jason X. -J. Yuan
- Departments of Medicine, COMRB Rm. 3131 (MC 719), University of Illinois at Chicago, 909 South Wolcott Avenue, Chicago, 60612 Illinois USA
| | - Joe G.N. Garcia
- 310 Admin.Office Building (MC 672), University of Illinois at Chicago, 1737 W. Polk Street, Suite 310, Chicago, 60612 Illinois USA
| | - John B. West
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093-0623 California USA
| | - Charles A. Hales
- Dept. Pulmonary & Critical Care Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, 02114 Massachusetts USA
| | - Stuart Rich
- Department of Medicine, University of Chicago Medical Center, 5841 S. Maryland Ave., Chicago, 60637 Illinois USA
| | - Stephen L. Archer
- Department of Medicine, University of Chicago School of Medicine, 5841 S. Maryland Ave., Chicago, 60637 Illinois USA
| |
Collapse
|
40
|
Mirzapoiazova T, Moitra J, Moreno-Vinasco L, Sammani S, Turner JR, Chiang ET, Evenoski C, Wang T, Singleton PA, Huang Y, Lussier YA, Watterson DM, Dudek SM, Garcia JGN. Non-muscle myosin light chain kinase isoform is a viable molecular target in acute inflammatory lung injury. Am J Respir Cell Mol Biol 2011; 44:40-52. [PMID: 20139351 PMCID: PMC3028257 DOI: 10.1165/rcmb.2009-0197oc] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Accepted: 11/24/2009] [Indexed: 01/03/2023] Open
Abstract
Acute lung injury (ALI) and mechanical ventilator-induced lung injury (VILI), major causes of acute respiratory failure with elevated morbidity and mortality, are characterized by significant pulmonary inflammation and alveolar/vascular barrier dysfunction. Previous studies highlighted the role of the non-muscle myosin light chain kinase isoform (nmMLCK) as an essential element of the inflammatory response, with variants in the MYLK gene that contribute to ALI susceptibility. To define nmMLCK involvement further in acute inflammatory syndromes, we used two murine models of inflammatory lung injury, induced by either an intratracheal administration of lipopolysaccharide (LPS model) or mechanical ventilation with increased tidal volumes (the VILI model). Intravenous delivery of the membrane-permeant MLC kinase peptide inhibitor, PIK, produced a dose-dependent attenuation of both LPS-induced lung inflammation and VILI (~50% reductions in alveolar/vascular permeability and leukocyte influx). Intravenous injections of nmMLCK silencing RNA, either directly or as cargo within angiotensin-converting enzyme (ACE) antibody-conjugated liposomes (to target the pulmonary vasculature selectively), decreased nmMLCK lung expression (∼70% reduction) and significantly attenuated LPS-induced and VILI-induced lung inflammation (∼40% reduction in bronchoalveolar lavage protein). Compared with wild-type mice, nmMLCK knockout mice were significantly protected from VILI, with significant reductions in VILI-induced gene expression in biological pathways such as nrf2-mediated oxidative stress, coagulation, p53-signaling, leukocyte extravasation, and IL-6-signaling. These studies validate nmMLCK as an attractive target for ameliorating the adverse effects of dysregulated lung inflammation.
Collapse
Affiliation(s)
- Tamara Mirzapoiazova
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Jaideep Moitra
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Liliana Moreno-Vinasco
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Saad Sammani
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Jerry R. Turner
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Eddie T. Chiang
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Carrie Evenoski
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Ting Wang
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Patrick A. Singleton
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Yong Huang
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Yves A. Lussier
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - D. Martin Watterson
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Steven M. Dudek
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| | - Joe G. N. Garcia
- Department of Medicine, University of Chicago; Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago; Department of Pathology; Section of Genetic Medicine, University of Chicago; Northwestern Medical School; and Institute for Personalized and Respiratory Medicine, University of Illinois at Chicago, Chicago Illinois
| |
Collapse
|
41
|
Acute Lung Injury: The Injured Lung Endothelium, Therapeutic Strategies for Barrier Protection, and Vascular Biomarkers. TEXTBOOK OF PULMONARY VASCULAR DISEASE 2010. [PMCID: PMC7120335 DOI: 10.1007/978-0-387-87429-6_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
42
|
Genetic variation in MYLK and lung injury in children and adults with community-acquired pneumonia. Pediatr Crit Care Med 2010; 11:731-6. [PMID: 20081554 DOI: 10.1097/pcc.0b013e3181ce7497] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate whether selected single nucleotide polymorphisms in the myosin light chain kinase gene are associated with more severe lung injury in children and adults with community-acquired pneumonia. Previous studies have demonstrated an association between single nucleotide polymorphisms in the myosin light chain kinase gene and increased severity of acute lung injury in adults. DESIGN Prospective, case-control genetic association study. SETTING Three tertiary children's hospitals and one adult healthcare system. PATIENTS A total of 800 pediatric patients and 393 adult patients. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Genetic variation in the myosin light chain kinase gene was examined. The pediatric cohort was predominantly composed of African American (n = 443) and Caucasian (n = 253) children. A total of 393 patients made up the adult cohort. Within the pediatric cohort, single nucleotide polymorphisms rs16834493, rs820463, and rs9840993 were genotyped in the African American patients, whereas single nucleotide polymorphisms rs960224, rs33264, rs11718105, and rs9289225 were genotyped in the Caucasian patients. One single nucleotide polymorphism (rs820336) was genotyped in both groups. Genotyping in the adult cohort included rs820336, rs860224, rs33264, and rs11718105. Genotyping was performed using the Taqman Assay. Data were analyzed separately for African Americans and Caucasians and for children and adults. No associations were observed between the myosin light chain kinase gene single nucleotide polymorphisms genotyped in children with community-acquired pneumonia and increased severity of lung injury. Similarly, no associations were observed between myosin light chain kinase gene single nucleotide polymorphisms genotyped in adults with community-acquired pneumonia and increased severity of lung injury. CONCLUSIONS No association between the selected single nucleotide polymorphisms in the myosin light chain kinase gene and either the need for positive-pressure ventilation or the development of acute lung injury/acute respiratory distress syndrome was observed in children with community-acquired pneumonia. This suggests that variation in this gene may play less of a role in lung injury in children or adults with community-acquired pneumonia than in adults with sepsis or trauma.
Collapse
|
43
|
Dudek SM, Chiang ET, Camp SM, Guo Y, Zhao J, Brown ME, Singleton PA, Wang L, Desai A, Arce FT, Lal R, Van Eyk JE, Imam SZ, Garcia JGN. Abl tyrosine kinase phosphorylates nonmuscle Myosin light chain kinase to regulate endothelial barrier function. Mol Biol Cell 2010; 21:4042-56. [PMID: 20861316 PMCID: PMC2982111 DOI: 10.1091/mbc.e09-10-0876] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
This study identified multiple novel c-Abl–mediated nmMLCK phosphorylation sites by mass spectroscopy and examined their influence on nmMLCK function and human lung endothelial barrier regulation. The data indicate an essential role for Abl kinase in vascular barrier regulation via phosphorylation of nmMLCK and the actin-binding protein cortactin. Nonmuscle myosin light chain kinase (nmMLCK), a multi-functional cytoskeletal protein critical to vascular homeostasis, is highly regulated by tyrosine phosphorylation. We identified multiple novel c-Abl–mediated nmMLCK phosphorylation sites by mass spectroscopy analysis (including Y231, Y464, Y556, Y846) and examined their influence on nmMLCK function and human lung endothelial cell (EC) barrier regulation. Tyrosine phosphorylation of nmMLCK increased kinase activity, reversed nmMLCK-mediated inhibition of Arp2/3-mediated actin polymerization, and enhanced binding to the critical actin-binding phosphotyrosine protein, cortactin. EC challenge with sphingosine 1-phosphate (S1P), a potent barrier-enhancing agonist, resulted in c-Abl and phosphorylated nmMLCK recruitment into caveolin-enriched microdomains, rapid increases in Abl kinase activity, and spatial targeting of c-Abl to barrier-promoting cortical actin structures. Conversely, reduced c-Abl expression in EC (siRNA) markedly attenuated S1P-mediated cortical actin formation, reduced the EC modulus of elasticity (assessed by atomic force microscopy), reduced nmMLCK and cortactin tyrosine phosphorylation, and attenuated S1P-mediated barrier enhancement. These studies indicate an essential role for Abl kinase in vascular barrier regulation via posttranslational modification of nmMLCK and strongly support c-Abl-cortactin-nmMLCK interaction as a novel determinant of cortical actin-based cytoskeletal rearrangement critical to S1P-mediated EC barrier enhancement.
Collapse
Affiliation(s)
- Steven M Dudek
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sun X, Ma SF, Wade MS, Flores C, Pino-Yanes M, Moitra J, Ober C, Kittles R, Husain AN, Ford JG, Garcia JGN. Functional variants of the sphingosine-1-phosphate receptor 1 gene associate with asthma susceptibility. J Allergy Clin Immunol 2010; 126:241-9, 249.e1-3. [PMID: 20624651 DOI: 10.1016/j.jaci.2010.04.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 03/15/2010] [Accepted: 04/23/2010] [Indexed: 01/16/2023]
Abstract
BACKGROUND The genetic mechanisms underlying asthma remain unclear. Increased permeability of the microvasculature is a feature of asthma, and the sphingosine-1-phosphate receptor (S1PR1) is an essential participant regulating lung vascular integrity and responses to lung inflammation. OBJECTIVE We explored the contribution of polymorphisms in the S1PR1 gene to asthma susceptibility. METHODS A combination of gene resequencing for single nucleotide polymorphism (SNP) discovery, case-control association, functional evaluation of associated SNPs, and protein immunochemistry studies was used. RESULTS Immunohistochemistry studies demonstrated significantly decreased S1PR1 protein expression in pulmonary vessels in lungs of asthmatic patients compared with those of nonasthmatic subjects (P < .05). Direct DNA sequencing of 27 multiethnic samples identified 39 S1PR1 variants (18 novel SNPs). Association studies were performed based on genotyping results from cosmopolitan tagging SNPs in 3 case-control cohorts from Chicago and New York totaling 1,061 subjects (502 cases and 559 control subjects). The promoter SNP rs2038366 (-1557G/T) was found to be associated with asthma (P = .03) in European Americans. In African Americans an association was found for both asthma and severe asthma for intronic SNP rs3753194 (c.-164+170A/G; P = .006 and P = .040, respectively) and for promoter SNP rs59317557 (-532C/G) with severe asthma (P = .028). Consistent with predicted in silico functionality, alleles of the promoter SNPs rs2038366 (-1557G/T) and rs59317557 (-532C/G) influenced the activity of a luciferase S1PR1 reporter vector in transfected endothelial cells exposed to growth factors (epidermal growth factor, platelet-derived growth factor, and vascular endothelial growth factor) known to be increased in asthmatic airways. CONCLUSION These data provide strong support for a role for S1PR1 gene variants in asthma susceptibility and severity.
Collapse
Affiliation(s)
- Xiaoguang Sun
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Klassert TE, Sánchez JJ, Almeida TA, Candenas L, Pinto F, Acosta O, Hernández M. Common variants of the neuropeptide expressing tachykinin genes and susceptibility to asthma: a case-control study. J Neuroimmunol 2010; 227:202-7. [PMID: 20580442 DOI: 10.1016/j.jneuroim.2010.05.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 05/21/2010] [Accepted: 05/26/2010] [Indexed: 01/01/2023]
Abstract
Since tachykinins appear to be involved in the pathogenesis of allergic asthma, we investigated a possible association between 28 single nucleotide polymorphisms of the tachykinin genes TAC1, TAC3 and TAC4, and asthma susceptibility. A case-control study was conducted on 102 patients and 100 healthy subjects from the Canary Islands (Spain). A significant association with asthma was observed for two SNPs: rs2291855 in the TAC3 gene conferring asthma protection (Odds ratio [OR]: 0.46; 95% Confidence Interval [CI]: 0.22-0.97; P=0.038), and rs4794068 in the TAC4 gene associated with an increased risk for asthma (OR: 1.94; 95% CI: 1.06-3.54; P=0.03). The present study represents a preliminary step in elucidating the association between tachykinin gene polymorphisms and asthma susceptibility.
Collapse
Affiliation(s)
- Tilman E Klassert
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez s/n, 38271 La Laguna, Canarias, Spain.
| | | | | | | | | | | | | |
Collapse
|
46
|
Kim SS, Kim JR, Moon JK, Choi BH, Kim TH, Kim KS, Kim JJ, Lee CK. Transcriptional alteration of p53 related processes as a key factor for skeletal muscle characteristics in Sus scrofa. Mol Cells 2009; 28:565-73. [PMID: 19937136 DOI: 10.1007/s10059-009-0159-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 09/22/2009] [Indexed: 12/14/2022] Open
Abstract
The pig could be a useful model to characterize molecular aspects determining several delicate phenotypes because they have been bred for those characteristics. The Korean native pig (KNP) is a regional breed in Korea that was characterized by relatively high intramuscular fat content and reddish meat color compared to other western breeds such as Yorkshire (YS). YS grew faster and contained more lean muscle than KNP. We compared the KNP to Yorksire to find molecular clues determining muscle characteristics. The comparison of skeletal gene expression profiles between these two breeds showed molecular differences in muscle. We found 82 differentially expressed genes (DEGs) defined by fold change (more than 1.5 fold difference) and statistical significance (within 5% of false discovery rate). Functional analyses of these DEGs indicated up-regulation of most genes involved in cell cycle arrest, down-regulation of most genes involved in cellular differentiation and its inhibition, down-regulation of most genes encoding component of muscular-structural system, and up-regulation of most genes involved in diverse metabolism in KNP. Especially, DEGs in above-mentioned categories included a large number of genes encoding proteins directly or indirectly involved in p53 pathway. Our results indicated a possible role of p53 to determine muscle characteristics between these two breeds.
Collapse
Affiliation(s)
- Seung-Soo Kim
- College of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee SO, Cheong HS, Park BL, Bae JS, Sim WC, Chun JY, Isbat M, Uh ST, Kim YH, Jang AS, Park CS, Shin HD. MYLK polymorphism associated with blood eosinophil level among asthmatic patients in a Korean population. Mol Cells 2009; 27:175-81. [PMID: 19277499 DOI: 10.1007/s10059-009-0022-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Revised: 11/04/2008] [Accepted: 11/25/2008] [Indexed: 10/24/2022] Open
Abstract
The myosin light chain kinase (MYLK) gene encodes both smooth muscle and nonmuscle cell isoforms. Recently, polymorphisms in MYLK have been reported to be associated with several diseases. To examine the genetic effects of polymorphisms on the risk of asthma and related phenotypes, we scrutinized MYLK by re-sequencing/genotyping and statistical analysis in Korean population (n = 1,015). Seventeen common polymorphisms located in or near exons, having pairwise r ( 2 ) values less than 0.25, were genotyped. Our statistical analysis did not replicate the associations with the risk of asthma and log-transformed total IgE levels observed among African descendant populations. However, two SNPs in intron 16 (+89872C > G and +92263T >C), which were in tight LD (|D'| = 0.99), revealed significant association with log-transformed blood eosinophil level even after correction multiple testing (P = 0.002/P( corr )= 0.01 and P = 0.002/P( corr ) = 0.01, respectively). The log-transformed blood eosinophil levels were higher in individuals bearing the minor alleles for +89872C > G and +92263T > C, than in those bearing other allele. In additional subgroup analysis, the genetic effects of both SNPs were much more apparent among asthmatic patients and atopic asthma patients. Among atopic asthma patients, the log-transformed blood eosinophil levels were proportionally increased by gene-dose dependent manner of in both +89872C > G and +92263T > C (P = 0.0002 and P = 0.00007, respectively). These findings suggest that MYLK polymorphisms might be among the genetic factors underlying differential increases of blood eosinophil levels among asthmatic patients. Further biological and/or functional studies are needed to confirm our results.
Collapse
Affiliation(s)
- Soo Ok Lee
- Department of Genetic Epidemiology, SNP Genetics, Inc., Seoul, 153-803, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ma SF, Flores C, Dudek SM, Nicolae DL, Ober C, Garcia JG. A common cortactin gene variation confers differential susceptibility to severe asthma. Genet Epidemiol 2008; 32:757-66. [PMID: 18521921 PMCID: PMC3774307 DOI: 10.1002/gepi.20343] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Genomic regions with replicated linkage to asthma-related phenotypes likely harbor multiple susceptibility loci with relatively minor effects on disease susceptibility. The 11q13 chromosomal region has repeatedly been linked to asthma with five genes residing in this region with reported replicated associations. Cortactin, an actin-binding protein encoded by the CTTN gene in 11q13, constitutes a key regulator of cytoskeletal dynamics and contractile cell machinery, events facilitated by interaction with myosin light chain kinase; encoded by MYLK, a gene we recently reported as associated with severe asthma in African Americans. To evaluate potential association of CTTN gene variation with asthma susceptibility, CTTN exons and flanking regions were re-sequenced in 48 non-asthmatic multiethnic samples, leading to selection of nine tagging polymorphisms for case-control association studies in individuals of European and African descent. After ancestry adjustments, an intronic variant (rs3802780) was significantly associated with severe asthma (odds ratio [OR]: 1.71; 95% confidence interval [CI]: 1.20-2.43; p=0.003) in a joint analysis. Further analyses evidenced independent and additive effects of CTTN and MYLK risk variants for severe asthma susceptibility in African Americans (accumulated OR: 2.93, 95% CI: 1.40-6.13, p=0.004). These data suggest that CTTN gene variation may contribute to severe asthma and that the combined effects of CTTN and MYLK risk polymorphisms may further increase susceptibility to severe asthma in African Americans harboring both genetic variants.
Collapse
Affiliation(s)
- Shwu-Fan Ma
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Carlos Flores
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Steven M. Dudek
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Dan L. Nicolae
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Joe G.N. Garcia
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
49
|
Variation in the myosin light chain kinase gene is associated with development of acute lung injury after major trauma. Crit Care Med 2008; 36:2794-800. [PMID: 18828194 DOI: 10.1097/ccm.0b013e318186b843] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Single nucleotide polymorphisms in the myosin light chain kinase (MYLK) gene have been implicated in the risk of sepsis-related acute lung injury and asthma. MYLK encodes protein isoforms involved in multiple components of the inflammatory response, including apoptosis, vascular permeability, and leukocyte diapedesis. We tested the association of MYLK gene variation in the development of acute lung injury in major trauma patients. METHODS We conducted a prospective cohort study of 273 subjects with major trauma (injury severity score > or = 16). All x-rays and clinical data were reviewed by three clinicians for acute lung injury classification. A total of 17 tagging single nucleotide polymorphisms in MYLK were genotyped. Single nucleotide polymorphisms were individually assessed at the genotype level, and multiple logistic regression models were used to adjust for baseline variables. Haplotype analyses of sliding windows including 2-5 single nucleotide polymorphisms were conducted. RESULTS Ninety-one of the 273 subjects (33%) met criteria for acute lung injury within 5 days of traumatic insult. Three informative MYLK coding single nucleotide polymorphisms were individually associated with acute lung injury, with two informative risk-conferring genotypes His21Pro (CC genotype, odds ratio = 1.87, 95% confidence interval 1.06-3.33; p = 0.022) and Pro147Ser (TT, odds ratio = 2.13, 95% confidence interval 1.14-4.10; p = 0.011) more frequent than the noninformative Thr335Thr CC genotype (odds ratio = 0.42, 95% confidence interval 0.20-0.85; p = 0.010). Each of these genotypic associations was more pronounced in African Americans with trauma. Multivariate analyses demonstrated the association of each MYLK single nucleotide polymorphism with acute lung injury to be independent of age, injury severity score, Acute Physiology and Chronic Health Evaluation III, and the mechanism of trauma. Finally, haplotype analyses revealed strong acute lung injury associations with 2-4 single nucleotide polymorphism haplotypes, all involving His21Pro (p < 0.008). CONCLUSIONS Three MYLK coding single nucleotide polymorphisms previously associated with sepsis-induced acute lung injury and severe asthma in African Americans were associated with acute lung injury development after trauma in African Americans, although effect directions differed. These results confirm our prior studies implicating MYLK as a susceptibility gene in a distinct acute lung injury subset other than sepsis.
Collapse
|
50
|
Miao H, Chen L, Riordan SM, Li W, Juarez S, Crabb AM, Lukas TJ, Du P, Lin SM, Wise A, Agapova OA, Yang P, Gu CC, Hernandez MR. Gene expression and functional studies of the optic nerve head astrocyte transcriptome from normal African Americans and Caucasian Americans donors. PLoS One 2008; 3:e2847. [PMID: 18716680 PMCID: PMC2518525 DOI: 10.1371/journal.pone.0002847] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Accepted: 07/07/2008] [Indexed: 11/19/2022] Open
Abstract
PURPOSE To determine whether optic nerve head (ONH) astrocytes, a key cellular component of glaucomatous neuropathy, exhibit differential gene expression in primary cultures of astrocytes from normal African American (AA) donors compared to astrocytes from normal Caucasian American (CA) donors. METHODS We used oligonucleotide Affymetrix microarray (HG U133A & HG U133A 2.0 chips) to compare gene expression levels in cultured ONH astrocytes from twelve CA and twelve AA normal age matched donor eyes. Chips were normalized with Robust Microarray Analysis (RMA) in R using Bioconductor. Significant differential gene expression levels were detected using mixed effects modeling and Statistical Analysis of Microarray (SAM). Functional analysis and Gene Ontology were used to classify differentially expressed genes. Differential gene expression was validated by quantitative real time RT-PCR. Protein levels were detected by Western blots and ELISA. Cell adhesion and migration assays tested physiological responses. Glutathione (GSH) assay detected levels of intracellular GSH. RESULTS Multiple analyses selected 87 genes differentially expressed between normal AA and CA (P<0.01). The most relevant genes expressed in AA were categorized by function, including: signal transduction, response to stress, ECM genes, migration and cell adhesion. CONCLUSIONS These data show that normal astrocytes from AA and CA normal donors display distinct expression profiles that impact astrocyte functions in the ONH. Our data suggests that differences in gene expression in ONH astrocytes may be specific to the development and/or progression of glaucoma in AA.
Collapse
Affiliation(s)
- Haixi Miao
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Lin Chen
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Sean M. Riordan
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Wenjun Li
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Santiago Juarez
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Andrea M. Crabb
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Thomas J. Lukas
- Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Pan Du
- Robert H, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Simon M. Lin
- Robert H, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Alexandria Wise
- Department of Biology, City College of New York, New York, New York, United States of America
| | - Olga A. Agapova
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ping Yang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Charles C. Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - M. Rosario Hernandez
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|