1
|
Johnson EE, Southern WM, Doud B, Steiger B, Razzoli M, Bartolomucci A, Ervasti JM. Retention of stress susceptibility in the mdx mouse model of Duchenne muscular dystrophy after PGC-1α overexpression or ablation of IDO1 or CD38. Hum Mol Genet 2024; 33:594-611. [PMID: 38181046 PMCID: PMC10954366 DOI: 10.1093/hmg/ddad206] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal degenerative muscle wasting disease caused by the loss of the structural protein dystrophin with secondary pathological manifestations including metabolic dysfunction, mood and behavioral disorders. In the mildly affected mdx mouse model of DMD, brief scruff stress causes inactivity, while more severe subordination stress results in lethality. Here, we investigated the kynurenine pathway of tryptophan degradation and the nicotinamide adenine dinucleotide (NAD+) metabolic pathway in mdx mice and their involvement as possible mediators of mdx stress-related pathology. We identified downregulation of the kynurenic acid shunt, a neuroprotective branch of the kynurenine pathway, in mdx skeletal muscle associated with attenuated peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) transcriptional regulatory activity. Restoring the kynurenic acid shunt by skeletal muscle-specific PGC-1α overexpression in mdx mice did not prevent scruff -induced inactivity, nor did abrogating extrahepatic kynurenine pathway activity by genetic deletion of the pathway rate-limiting enzyme, indoleamine oxygenase 1. We further show that reduced NAD+ production in mdx skeletal muscle after subordination stress exposure corresponded with elevated levels of NAD+ catabolites produced by ectoenzyme cluster of differentiation 38 (CD38) that have been implicated in lethal mdx response to pharmacological β-adrenergic receptor agonism. However, genetic CD38 ablation did not prevent mdx scruff-induced inactivity. Our data do not support a direct contribution by the kynurenine pathway or CD38 metabolic dysfunction to the exaggerated stress response of mdx mice.
Collapse
Affiliation(s)
- Erynn E Johnson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, 420 Delaware St. SE, Minneapolis, MN 55455, United States
| | - W Michael Southern
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, 420 Delaware St. SE, Minneapolis, MN 55455, United States
| | - Baird Doud
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, 420 Delaware St. SE, Minneapolis, MN 55455, United States
| | - Brandon Steiger
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, 420 Delaware St. SE, Minneapolis, MN 55455, United States
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 321 Church St. SE, Minneapolis, MN 55455, United States
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 321 Church St. SE, Minneapolis, MN 55455, United States
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, 420 Delaware St. SE, Minneapolis, MN 55455, United States
| |
Collapse
|
2
|
Guerrero SC, Panettieri RA, Rastogi D. Mechanistic Links Between Obesity and Airway Pathobiology Inform Therapies for Obesity-Related Asthma. Paediatr Drugs 2023; 25:283-299. [PMID: 36656428 PMCID: PMC11071627 DOI: 10.1007/s40272-022-00554-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2022] [Indexed: 01/20/2023]
Abstract
Obesity-related asthma is associated with a high disease burden and a poor response to existent asthma therapies, suggesting that it is a distinct asthma phenotype. The proposed mechanisms that contribute to obesity-related asthma include the effects of the mechanical load of obesity, adipokine perturbations, and immune dysregulation. Each of these influences airway smooth muscle function. Mechanical fat load alters airway smooth muscle stretch affecting airway wall geometry, airway smooth muscle contractility, and agonist delivery; weight loss strategies, including medically induced weight loss, counter these effects. Among the metabolic disturbances, insulin resistance and free fatty acid receptor activation influence distinct signaling pathways in the airway smooth muscle downstream of both the M2 muscarinic receptor and the β2 adrenergic receptor, such as phospholipase C and the extracellular signal-regulated kinase signaling cascade. Medications that decrease insulin resistance and dyslipidemia are associated with a lower asthma disease burden. Leptin resistance is best understood to modulate muscarinic receptors via the neural pathways but there are no specific therapies for leptin resistance. From the immune perspective, monocytes and T helper cells are involved in systemic pro-inflammatory profiles driven by obesity, notably associated with elevated levels of interleukin-6. Clinical trials on tocilizumab, an anti-interleukin antibody, are ongoing for obesity-related asthma. This armamentarium of therapies is distinct from standard asthma medications, and once investigated for its efficacy and safety among children, will serve as a novel therapeutic intervention for pediatric obesity-related asthma. Irrespective of the directionality of the association between asthma and obesity, airway-specific mechanistic studies are needed to identify additional novel therapeutic targets for obesity-related asthma.
Collapse
Affiliation(s)
- Silvia Cabrera Guerrero
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, 111 Michigan Ave NW, Washington, DC, 20010, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Deepa Rastogi
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, 111 Michigan Ave NW, Washington, DC, 20010, USA.
| |
Collapse
|
3
|
Maples KT, Hall KH, Joseph NS, Hofmeister CC, Gupta V, Dhodapkar MV, Kaufman JL, Nooka AK, Lonial S. Eliminating the monitoring period with subcutaneous daratumumab: a single-center experience. Blood Cancer J 2023; 13:29. [PMID: 36804376 PMCID: PMC9939853 DOI: 10.1038/s41408-023-00801-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Affiliation(s)
- Kathryn T Maples
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA.
| | - Kevin H Hall
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Nisha S Joseph
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Vikas Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
The Role of CD38 in the Pathogenesis of Cardiorenal Metabolic Disease and Aging, an Approach from Basic Research. Cells 2023; 12:cells12040595. [PMID: 36831262 PMCID: PMC9954496 DOI: 10.3390/cells12040595] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Aging is a major risk factor for the leading causes of mortality, and the incidence of age-related diseases including cardiovascular disease, kidney disease and metabolic disease increases with age. NAD+ is a classic coenzyme that exists in all species, and that plays a crucial role in oxidation-reduction reactions. It is also involved in the regulation of many cellular functions including inflammation, oxidative stress and differentiation. NAD+ declines with aging in various organs, and the reduction in NAD+ is possibly involved in the development of age-related cellular dysfunction in cardiorenal metabolic organs through the accumulation of inflammation and oxidative stress. Levels of NAD+ are regulated by the balance between its synthesis and degradation. CD38 is the main NAD+-degrading enzyme, and CD38 is activated in response to inflammation with aging, which is associated with the reduction in NAD+ levels. In this review, focusing on CD38, we discuss the role of CD38 in aging and the pathogenesis of age-related diseases, including cardiorenal metabolic disease.
Collapse
|
5
|
Carvalho de Oliveira J, Mathias C, Oliveira VC, Pezuk JA, Brassesco MS. The Double Face of miR-708: A Pan-Cancer Player with Dissociative Identity Disorder. Genes (Basel) 2022; 13:genes13122375. [PMID: 36553642 PMCID: PMC9777992 DOI: 10.3390/genes13122375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Over the last decades, accumulating evidence has shown tumor-dependent profiles of miR-708, being either up- or downregulated, and thus, acting as a "Janus" regulator of oncogenic pathways. Herein, its functional duality was assessed through a thorough review of the literature and further validation in silico using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. In the literature, miR-708 was found with an oncogenic role in eight tumor types, while a suppressor tumor role was described in seven cancers. This double profile was also found in TCGA and GEO databases, with some tumor types having a high expression of miR-708 and others with low expression compared with non-tumor counterparts. The investigation of validated targets using miRBase, miRTarBase, and miRecords platforms, identified a total of 572 genes that appeared enriched for PI3K-Akt signaling, followed by cell cycle control, p53, Apellin and Hippo signaling, endocrine resistance, focal adhesion, and cell senescence regulations, which are all recognized contributors of tumoral phenotypes. Among these targets, a set of 15 genes shared by at least two platforms was identified, most of which have important roles in cancer cells that influence either tumor suppression or progression. In a clinical scenario, miR-708 has shown to be a good diagnostic and prognosis marker. However, its multitarget nature and opposing roles in diverse human tumors, aligned with insufficient experimental data and the lack of proper delivery strategies, hamper its potential as a sequence-directed therapeutic.
Collapse
Affiliation(s)
| | - Carolina Mathias
- Department of Genetics, Federal University of Paraná, Curitiba 80060-000, Brazil
- Laboratory of Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, Brazil
| | - Verônica Cristina Oliveira
- Department of Biotechnology and Health Innovation, Anhanguera University of São Paulo, Pirituba 05145-200, Brazil
| | - Julia Alejandra Pezuk
- Department of Biotechnology and Health Innovation, Anhanguera University of São Paulo, Pirituba 05145-200, Brazil
| | - María Sol Brassesco
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, Brazil
- Correspondence:
| |
Collapse
|
6
|
Okwuofu EO, Hui AYC, Woei JLC, Stanslas J. Molecular and Immunomodulatory Actions of New Antiasthmatic Agents: Exploring the Diversity of Biologics in Th2 Endotype Asthma. Pharmacol Res 2022; 181:106280. [PMID: 35661709 DOI: 10.1016/j.phrs.2022.106280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/17/2022] [Accepted: 05/26/2022] [Indexed: 02/07/2023]
Abstract
Asthma is a major respiratory disorder characterised by chronic inflammation and airway remodelling. It affects about 1-8% of the global population and is responsible for over 461,000 deaths annually. Until recently, the pharmacotherapy of severe asthma involved high doses of inhaled corticosteroids in combination with β-agonist for prolonged action, including theophylline, leukotriene antagonist or anticholinergic yielding limited benefit. Although the use of newer agents to target Th2 asthma endotypes has improved therapeutic outcomes in severe asthmatic conditions, there seems to be a paucity of understanding the diverse mechanisms through which these classes of drugs act. This article delineates the molecular and immunomodulatory mechanisms of action of new antiasthmatic agents currently being trialled in preclinical and clinical studies to remit asthmatic conditions. The ultimate goal in developing antiasthmatic agents is based on two types of approaches: either anti-inflammatory or bronchodilators. Biologic and most small molecules have been shown to modulate specific asthma endotypes, targeting thymic stromal lymphopoietin, tryptase, spleen tyrosine kinase (Syk), Janus kinase, PD-L1/PD-L2, GATA-3, and CD38 for the treatment and management of Th2 endotype asthma.
Collapse
Affiliation(s)
- Emmanuel Oshiogwe Okwuofu
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | - Jonathan Lim Chee Woei
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|
7
|
Huang ZX, Qiu ZE, Chen L, Hou XC, Zhu YX, Zhou WL, Zhang YL. Cellular mechanism underlying the facilitation of contractile response induced by IL-25 in mouse tracheal smooth muscle. Am J Physiol Lung Cell Mol Physiol 2022; 323:L27-L36. [PMID: 35537103 DOI: 10.1152/ajplung.00468.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Asthma is a common heterogeneous respiratory disease characterized by airway inflammation and airway hyperresponsiveness (AHR) which is associated with abnormality in smooth muscle contractility. The epithelial cell-derived cytokine IL-25 is implicated in type 2 immune pathology including asthma, whereas the underlying mechanisms have not been fully elucidated. This study aims to investigate the effects of IL-25 on mouse tracheal smooth muscle contractility and elucidate the cellular mechanisms. Incubation with IL-25 augmented the contraction of mouse tracheal smooth muscles, which could be suppressed by the L-type voltage-dependent Ca2+ channel (L-VDCC) blocker nifedipine. Furthermore, IL-25 enhanced the cytosolic Ca2+ signals and triggered up-regulation of α1C L-VDCC (CaV1.2) in primary cultured mouse tracheal smooth muscle cells. Knocking down IL-17RA/IL-17RB receptors or inhibiting the transforming growth factor-β-activated kinase 1 (TAK1)-tumor progression locus 2 (TPL2)-MAPK kinase 1/2 (MEK1/2)-ERK1/2-activating protein-1 (AP-1) signaling pathways suppressed the IL-25-elicited up-regulation of CaV1.2 and hyperreactivity in tracheal smooth muscles. Moreover, inhibition of TPL2, ERK1/2 or L-VDCC alleviated the AHR symptom induced by IL-25 in a murine model. This study revealed that IL-25 potentiated the contraction of tracheal smooth muscle and evoked AHR via activation of TPL2-ERK1/2-CaV1.2 signaling, providing novel targets for the treatment of asthma with a high-IL-25 phenotype.
Collapse
Affiliation(s)
- Ze-Xin Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhuo-Er Qiu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Chun Hou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yun-Xin Zhu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Kim JH, Park SS, Yoon JH, Lee SE, Kim HJ, Min CK. Daratumumab monotherapy in relapsed and refractory multiple myeloma patients with severely compromised forced expiratory volume in one second. Blood Res 2022; 57:76-80. [PMID: 35342045 PMCID: PMC8958367 DOI: 10.5045/br.2022.2021183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/05/2021] [Accepted: 12/29/2021] [Indexed: 11/22/2022] Open
Affiliation(s)
- Jin-Hyo Kim
- Department of Hematology, Seoul St. Mary's Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Soo Park
- Department of Hematology, Seoul St. Mary's Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae-Ho Yoon
- Department of Hematology, Seoul St. Mary's Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Eun Lee
- Department of Hematology, Seoul St. Mary's Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee-Je Kim
- Department of Hematology, Seoul St. Mary's Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang-Ki Min
- Department of Hematology, Seoul St. Mary's Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
9
|
de Zélicourt A, Fayssoil A, Dakouane-Giudicelli M, De Jesus I, Karoui A, Zarrouki F, Lefebvre F, Mansart A, Launay JM, Piquereau J, Tarragó MG, Bonay M, Forand A, Moog S, Piétri-Rouxel F, Brisebard E, Chini CCS, Kashyap S, Fogarty MJ, Sieck GC, Mericskay M, Chini EN, Gomez AM, Cancela JM, de la Porte S. CD38-NADase is a new major contributor to Duchenne muscular dystrophic phenotype. EMBO Mol Med 2022; 14:e12860. [PMID: 35298089 PMCID: PMC9081905 DOI: 10.15252/emmm.202012860] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 01/14/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration. Two important deleterious features are a Ca2+ dysregulation linked to Ca2+ influxes associated with ryanodine receptor hyperactivation, and a muscular nicotinamide adenine dinucleotide (NAD+) deficit. Here, we identified that deletion in mdx mice of CD38, a NAD+ glycohydrolase‐producing modulators of Ca2+ signaling, led to a fully restored heart function and structure, with skeletal muscle performance improvements, associated with a reduction in inflammation and senescence markers. Muscle NAD+ levels were also fully restored, while the levels of the two main products of CD38, nicotinamide and ADP‐ribose, were reduced, in heart, diaphragm, and limb. In cardiomyocytes from mdx/CD38−/− mice, the pathological spontaneous Ca2+ activity was reduced, as well as in myotubes from DMD patients treated with isatuximab (SARCLISA®) a monoclonal anti‐CD38 antibody. Finally, treatment of mdx and utrophin–dystrophin‐deficient (mdx/utr−/−) mice with CD38 inhibitors resulted in improved skeletal muscle performances. Thus, we demonstrate that CD38 actively contributes to DMD physiopathology. We propose that a selective anti‐CD38 therapeutic intervention could be highly relevant to develop for DMD patients.
Collapse
Affiliation(s)
- Antoine de Zélicourt
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, Versailles, France.,Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, Saclay, France
| | | | | | - Isley De Jesus
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, Versailles, France
| | - Ahmed Karoui
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - Faouzi Zarrouki
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, Versailles, France
| | - Florence Lefebvre
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - Arnaud Mansart
- Université Paris-Saclay, UVSQ, Inserm, 2I, Versailles, France
| | - Jean-Marie Launay
- Service de Biochimie, INSERM UMR S942, Hôpital Lariboisière, Paris, France
| | - Jerome Piquereau
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - Mariana G Tarragó
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Marcel Bonay
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, Versailles, France
| | - Anne Forand
- Centre de Recherche en Myologie, Faculté de Médecine de la Pitié Salpêtrière, Sorbonne Université-UMRS974-Inserm-Institut de Myologie, Paris, France.,Inovarion, Paris, France
| | - Sophie Moog
- Centre de Recherche en Myologie, Faculté de Médecine de la Pitié Salpêtrière, Sorbonne Université-UMRS974-Inserm-Institut de Myologie, Paris, France.,Inovarion, Paris, France
| | - France Piétri-Rouxel
- Centre de Recherche en Myologie, Faculté de Médecine de la Pitié Salpêtrière, Sorbonne Université-UMRS974-Inserm-Institut de Myologie, Paris, France
| | | | - Claudia C S Chini
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonu Kashyap
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew J Fogarty
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Gary C Sieck
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Mathias Mericskay
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - Eduardo N Chini
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Ana Maria Gomez
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - José-Manuel Cancela
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, Saclay, France
| | | |
Collapse
|
10
|
Monoclonal Antibodies in Myeloma: Optimizing Targeted Therapy. Cancer J 2021; 27:222-230. [PMID: 34549911 DOI: 10.1097/ppo.0000000000000521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT In the past several years, there have been significant advances in the therapeutic arsenal of agents used to treat multiple myeloma (MM). Despite these advances, MM remains incurable. One of the most recent therapeutic advances is the development of targeted monoclonal antibodies (MoAbs). The MoAbs have significantly improved disease response rates, and extended survival in MM patients. In this review, we highlight the current US Food and Drug Administration approved MoAbs, namely, belantamab mafodotin, daratumumab, elotuzumab, and isatuximab. The mechanisms of action and pivotal clinical trials that led to US Food and Drug Administration approval of these agents and their current therapeutic use in the management of patients with MM are discussed in detail. Lastly, we describe several novel MoAbs under clinical investigation with potential for approval in the future.
Collapse
|
11
|
Gan L, Liu D, Liu J, Chen E, Chen C, Liu L, Hu H, Guan X, Ma W, Zhang Y, He Y, Liu B, Tang S, Jiang W, Xue J, Xin H. CD38 deficiency alleviates Ang II-induced vascular remodeling by inhibiting small extracellular vesicle-mediated vascular smooth muscle cell senescence in mice. Signal Transduct Target Ther 2021; 6:223. [PMID: 34112762 PMCID: PMC8192533 DOI: 10.1038/s41392-021-00625-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 03/29/2021] [Accepted: 04/27/2021] [Indexed: 02/05/2023] Open
Abstract
CD38 is the main enzyme for nicotinamide adenine dinucleotide (NAD) degradation in mammalian cells. Decreased NAD levels are closely related to metabolic syndromes and aging-related diseases. Our study showed that CD38 deficiency significantly alleviated angiotensin II (Ang II)-induced vascular remodeling in mice, as shown by decreased blood pressures; reduced vascular media thickness, media-to-lumen ratio, and collagen deposition; and restored elastin expression. However, our bone marrow transplantation assay showed that CD38 deficiency in lymphocytes led to lack of protection against Ang II-induced vascular remodeling, suggesting that the effects of CD38 on Ang II-induced vascular remodeling might rely primarily on vascular smooth muscle cells (VSMCs), not lymphocytes. In addition, we observed that CD38 deficiency or NAD supplementation remarkably mitigated Ang II-induced vascular senescence by suppressing the biogenesis, secretion, and internalization of senescence-associated small extracellular vesicles (SA-sEVs), which facilitated the senescence of neighboring non-damaged VSMCs. Furthermore, we found that the protective effects of CD38 deficiency on VSMC senescence were related to restoration of lysosome dysfunction, particularly with respect to the maintenance of sirtuin-mediated mitochondrial homeostasis and activation of the mitochondria-lysosomal axis in VSMCs. In conclusion, our findings demonstrated that CD38 and its associated intracellular NAD decline are critical for Ang II-induced VSMC senescence and vascular remodeling.
Collapse
Affiliation(s)
- Lu Gan
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Demin Liu
- Cardiology Department, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jing Liu
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Erya Chen
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chan Chen
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lian Liu
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hang Hu
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaohui Guan
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Wen Ma
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yanzi Zhang
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yarong He
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Bofu Liu
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Songling Tang
- Research Laboratory of Emergency Medicine, Department of Emergency Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Wei Jiang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jianxin Xue
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Hongbo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, People's Republic of China.
| |
Collapse
|
12
|
Jiang T, Li Z, Zhao D, Hui B, Zheng Z. SOX18 enhances the proliferation and migration of airway smooth muscle cells induced by tumor necrosis factor-α via the regulation of Notch1 signaling. Int Immunopharmacol 2021; 96:107746. [PMID: 34004439 DOI: 10.1016/j.intimp.2021.107746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/15/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023]
Abstract
Childhood asthma is a frequent chronic disease of pediatric populations. The excessive proliferation and migration of airway smooth muscle cells contribute to airway remodeling during asthma pathogenesis. Sex-determining region on the Y chromosome-related high mobility group box 18 (SOX18) has been reported to be over-expressed in asthma. However, whether SOX18 plays a role in modulating the airway remodeling of asthma is not fully understood. The purposes of this work were to assess the potential role of SOX18 in modulating airway remodeling using tumor necrosis factor-α (TNF-α)-stimulated airway smooth muscle cells in vitro. Our results showed that SOX18 expression was increased following TNF-α stimulation in airway smooth muscle cells. The silencing of SOX18 markedly prohibited the proliferation and migration of airway smooth muscle cells induced by TNF-α, whilst the over-expression of SOX18 produced the opposite effects. Further investigation revealed that SOX18 promoted the expression of Notch1, and enhanced the activation of Notch1 signaling in airway smooth muscle cells stimulated by TNF-α. The inhibition of Notch1 markedly diminished SOX18-over-expression-evoked promotion effects on TNF-α-induced proliferation and migration of airway smooth muscle cells. In addition, the reactivation of Notch1 signaling markedly reversed the SOX18-silencing-induced suppressive effect on the TNF-α-induced proliferation and the migration of airway smooth muscle cells. In summary, the findings of this work demonstrate that SOX18 regulates the proliferation and migration of airway smooth muscle cells induced by TNF-α via the modulation of Notch1 signaling. This study indicates a potential role for SOX18 in promoting airway remodeling during asthma pathogenesis.
Collapse
Affiliation(s)
- Te Jiang
- Pediatrics, Northwest Women's and Children's Hospital, Xi'an 610113, China
| | - Zhankui Li
- Pediatrics, Northwest Women's and Children's Hospital, Xi'an 610113, China.
| | - Di Zhao
- Pediatrics, Northwest Women's and Children's Hospital, Xi'an 610113, China
| | - Bengang Hui
- Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, China
| | - Zhiyuan Zheng
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University and Shanghai Institute of Medical Imaging, Shanghai 200032, China
| |
Collapse
|
13
|
Borkar NA, Roos B, Prakash YS, Sathish V, Pabelick CM. Nicotinic α7 acetylcholine receptor (α7nAChR) in human airway smooth muscle. Arch Biochem Biophys 2021; 706:108897. [PMID: 34004182 DOI: 10.1016/j.abb.2021.108897] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/31/2022]
Abstract
Diseases such as asthma are exacerbated by inflammation, cigarette smoke and even nicotine delivery devices such as e-cigarettes. However, there is currently little information on how nicotine affects airways, particularly in humans, and changes in the context of inflammation or asthma. Here, a longstanding assumption is that airway smooth muscle (ASM) that is key to bronchoconstriction has muscarinic receptors while nicotinic receptors (nAChRs) are only on airway neurons. In this study, we tested the hypothesis that human ASM expresses α7nAChR and explored its profile in inflammation and asthma using ASM of non-asthmatics vs. mild-moderate asthmatics. mRNA and western analysis showed the α7 subunit is most expressed in ASM cells and further increased in asthmatics and smokers, or by exposure to nicotine, cigarette smoke or pro-inflammatory cytokines TNFα and IL-13. In these effects, signaling pathways relevant to asthma such as NFκB, AP-1 and CREB are involved. These novel data demonstrate the expression of α7nAChR in human ASM and suggest their potential role in asthma pathophysiology in the context of nicotine exposure.
Collapse
Affiliation(s)
- Niyati A Borkar
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Benjamin Roos
- Department of Anesthesiology and Perioperative Medicine, USA
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
14
|
Richter J, Thibaud S. Anti-body building: The exercise of advancing immune based myeloma therapies. Blood Rev 2020; 48:100789. [PMID: 33384171 DOI: 10.1016/j.blre.2020.100789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 12/26/2022]
Abstract
The last decade has seen a marked improvement in the outcomes of patients with multiple myeloma. Much of this has been due to not only the advent of new therapies, but their inherent ability to be combined into 3 and 4 drug regimens without resulting in unacceptable toxicity. The general gestalt has been to combine agents of varied mechanisms of action. With the primary classes of agents such as proteasome inhibitors and immunomodulatory drugs as bases, the advent of antibody-based therapy in myeloma has allowed us to easily augment these therapies; much in the same way rituximab impacted the lymphoma world. With the approvals of daratumumab, elotuzumab and isatuximab; the myeloma world was ushered into the next wave of targeted agents. Here, we take a look at the current landscape of "off-the-shelf" antibody-based therapies in myeloma and peer into the next wave of multi-functional targeted agents.
Collapse
Affiliation(s)
- Joshua Richter
- Tisch Cancer Institute: Icahn School of Medicine at Mount Sinai, USA.
| | - Santiago Thibaud
- Tisch Cancer Institute: Icahn School of Medicine at Mount Sinai, USA.
| |
Collapse
|
15
|
Graeff R, Guedes A, Quintana R, Wendt-Hornickle E, Baldo C, Walseth T, O’Grady S, Kannan M. Novel Pathway of Adenosine Generation in the Lungs from NAD +: Relevance to Allergic Airway Disease. Molecules 2020; 25:molecules25214966. [PMID: 33120985 PMCID: PMC7663290 DOI: 10.3390/molecules25214966] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/24/2020] [Accepted: 10/26/2020] [Indexed: 11/20/2022] Open
Abstract
Adenosine and uric acid (UA) play a pivotal role in lung diseases such as asthma and chronic obstructive pulmonary disease (COPD). In the present experiments, we measured adenosine synthesis from nicotinamide adenine dinucleotide (NAD+) in membranes prepared from wild type (WT) and CD38 knockout (CD38KO) mouse lungs, from cultured airway smooth muscle and epithelial cells, and in bronchoalveolar lavage fluid after airway challenge with epidemiologically relevant allergens. Adenosine was determined using an enzymatically coupled assay that produces ATP and is detected by luminescence. Uric acid was determined by ELISA. Exposure of cultured airway epithelial cells to Alternaria alternata extract caused significant nucleotide (NAD+ and ATP) release in the culture media. The addition of NAD+ to membranes prepared from WT mice resulted in faster generation of adenosine compared to membranes from CD38KO mice. Formation of adenosine from NAD+ affected UA and ATP concentrations, its main downstream molecules. Furthermore, NAD+ and adenosine concentrations in the bronchoalveolar lavage fluid decreased significantly following airway challenge with house-dust mite extract in WT but not in CD38KO mice. Thus, NAD+ is a significant source of adenosine and UA in the airways in mouse models of allergic airway disease, and the capacity for their generation from NAD+ is augmented by CD38, a major NADase with high affinity for NAD+. This novel non-canonical NAD+-adenosine-UA pathway that is triggered by allergens has not been previously described in the airways.
Collapse
Affiliation(s)
- Richard Graeff
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA;
| | - Alonso Guedes
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (A.G.); (R.Q.); (E.W.-H.); (C.B.)
| | - Ruth Quintana
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (A.G.); (R.Q.); (E.W.-H.); (C.B.)
| | - Erin Wendt-Hornickle
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (A.G.); (R.Q.); (E.W.-H.); (C.B.)
| | - Caroline Baldo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (A.G.); (R.Q.); (E.W.-H.); (C.B.)
| | - Timothy Walseth
- Department of Pharmacology, University of Minnesota Medical School, University of Minnesota, St. Paul, MN 55455, USA;
| | - Scott O’Grady
- Department of Animal Science, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, St. Paul, MN 55108, USA;
| | - Mathur Kannan
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA;
- Correspondence:
| |
Collapse
|
16
|
Chiba Y, Matsumoto M, Hanazaki M, Sakai H. Downregulation of miR-140-3p Contributes to Upregulation of CD38 Protein in Bronchial Smooth Muscle Cells. Int J Mol Sci 2020; 21:E7982. [PMID: 33121100 PMCID: PMC7663226 DOI: 10.3390/ijms21217982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
In allergic bronchial asthma, an increased smooth muscle contractility of the airways is one of the causes of the airway hyperresponsiveness (AHR). Increasing evidence also suggests a possible involvement of microRNAs (miRNAs) in airway diseases, including asthma, although their roles in function and pathology largely unknown. The current study aimed to determine the role of a miRNA, miR-140-3p, in the control of protein expression of CD38, which is believed to regulate the contraction of smooth muscles, including the airways. In bronchial smooth muscles (BSMs) of the mice that were actively sensitized and repeatedly challenged with ovalbumin antigen, an upregulation of CD38 protein concurrently with a significant reduction of miR-140-3p was observed. In cultured human BSM cells (hBSMCs), transfection with a synthetic miR-140-3p inhibitor caused an increase in CD38 protein, indicating that its basal protein expression is regulated by endogenous miR-140-3p. Treatment of the hBSMCs with interleukin-13 (IL-13), an asthma-related cytokine, caused both an upregulation of CD38 protein and a downregulation of miR-140-3p. Transfection of the hBSMCs with miR-140-3p mimic inhibited the CD38 protein upregulation induced by IL-13. On the other hand, neither a CD38 product cyclic ADP-ribose (cADPR) nor its antagonist 8-bromo-cADPR had an effect on the BSM contraction even in the antigen-challenged mice. Taken together, the current findings suggest that the downregulation of miR-140-3p induced by IL-13 might cause an upregulation of CD38 protein in BSM cells of the disease, although functional and pathological roles of the upregulated CD38 are still unclear.
Collapse
Affiliation(s)
- Yoshihiko Chiba
- Laboratory of Molecular Biology and Physiology, School of Pharmacy, Hoshi University, Tokyo 142-8501, Japan; (M.M.); (M.H.)
| | - Mayumi Matsumoto
- Laboratory of Molecular Biology and Physiology, School of Pharmacy, Hoshi University, Tokyo 142-8501, Japan; (M.M.); (M.H.)
| | - Motohiko Hanazaki
- Laboratory of Molecular Biology and Physiology, School of Pharmacy, Hoshi University, Tokyo 142-8501, Japan; (M.M.); (M.H.)
- Department of Anesthesiology and Intensive Care Medicine, School of Medicine, International University of Health and Welfare, Chiba 286-8686, Japan
| | - Hiroyasu Sakai
- Laboratory of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, Tokyo 142-8501, Japan;
| |
Collapse
|
17
|
Wu Y, Lu Y, Zou F, Fan X, Li X, Zhang H, Chen H, Sun X, Liu Y. PTEN participates in airway remodeling of asthma by regulating CD38/Ca 2+/CREB signaling. Aging (Albany NY) 2020; 12:16326-16340. [PMID: 32889801 PMCID: PMC7485701 DOI: 10.18632/aging.103664] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022]
Abstract
Both phosphatase and tensin homologue deleted on chromosome ten (PTEN) and cluster of differentiation 38 (CD38) have been suggested to be key regulators of the pathogenesis of asthma. However, the precise role and molecular mechanisms by which PTEN and CD38 are involved in airway remodeling throughout asthma pathogenesis remains poorly understood. This study aimed to elucidate the role of PTEN and CD38 in airway remodeling of asthma. Exposure to tumor necrosis factor-α (TNF-α) in airway smooth muscle (ASM) cells markedly decreased PTEN expression, and increased expression of CD38. Overexpression of PTEN suppressed the expression of CD38 and downregulated proliferation and migration induced by TNF-α stimulation, which was partially reversed by CD38 overexpression. PTEN/CD38 axis regulated Ca2+ levels and cyclic AMP response-element binding protein (CREB) phosphorylation in TNF-α-stimulated ASM cells. The in vitro knockdown of CD38 or overexpression of PTEN remarkably restricted airway remodeling and decreased Ca2+ concentrations and CREB phosphorylation in asthmatic mice. CD38 overexpression abolished the inhibitory effects of PTEN overexpression on airway remodeling. These findings demonstrate that PTEN inhibits airway remodeling of asthma through the downregulation of CD38-mediated Ca2+/CREB signaling, highlighting a key role of PTEN/CD38/Ca2+/CREB signaling in the molecular pathogenesis of asthma.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Yiyi Lu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Fan Zou
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Xinping Fan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Xudong Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Hongni Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Haijuan Chen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Xiuzhen Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Yun Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| |
Collapse
|
18
|
Sowers ML, Tang H, Tian B, Goldblum R, Midoro-Horiuti T, Zhang K. Bisphenol A Activates an Innate Viral Immune Response Pathway. J Proteome Res 2020; 19:644-654. [PMID: 31816243 PMCID: PMC8311900 DOI: 10.1021/acs.jproteome.9b00548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bisphenol A (BPA) is a ubiquitous component in the manufacturing of plastic. It is commonly found in food and beverage containers. Because of its broad exposure and evidence that it may act as an estrogen-like molecule, many have studied its potential effects. For example, epidemiological studies have found an association between in utero BPA exposure and onset of childhood asthma. Our previous work suggested BPA treated mice induced asthma-like symptoms in both mothers and their pups. In order to better understand theconsequences of BPA exposure and potential mechanisms, we used a proteomics approach. Using both CD4+ T cells from an in vivo model of BPA exposure and an in vitro epithelial cell model, we identified activation of both innate and adaptive immune signaling following BPA exposure. Furthermore, our proteomic results from our multigenerational mouse model study implicates aberrant immune activation across several generations. We propose the following; BPA can active an innate viral immune response by upregulating a probable palmitoyltransferase ZDHHC1, and its binding partner stimulator of interferon-gamma (STING). It also has additional histone epigenetic perturbations, suggesting a role for epigenetic inheritance of these immune perturbations.
Collapse
Affiliation(s)
- Mark L. Sowers
- MD-PhD Combined Degree Program, University of Texas Medical Branch Galveston, Texas, 77555
- Department of Pharmacology and Toxicology, University of Texas Medical Branch Galveston, Texas, 77555
| | - Hui Tang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch Galveston, Texas, 77555
| | - Bing Tian
- Department of Internal Medicine-Endocrinology, University of Texas Medical Branch Galveston, Texas, 77555
| | - Randall Goldblum
- Department of Pediatrics Child Health Research Center, University of Texas Medical Branch Galveston, Texas, 77555
| | - Terumi Midoro-Horiuti
- Department of Pediatrics Child Health Research Center, University of Texas Medical Branch Galveston, Texas, 77555
| | - Kangling Zhang
- MD-PhD Combined Degree Program, University of Texas Medical Branch Galveston, Texas, 77555
| |
Collapse
|
19
|
SETD7 promotes TNF-α-induced proliferation and migration of airway smooth muscle cells in vitro through enhancing NF-κB/CD38 signaling. Int Immunopharmacol 2020; 72:459-466. [PMID: 31035088 DOI: 10.1016/j.intimp.2019.04.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/27/2019] [Accepted: 04/19/2019] [Indexed: 11/21/2022]
Abstract
The inflammation-induced the excessive proliferation and migration of airway smooth muscle (ASM) cells in the airway wall contribute to airway remodeling in asthma pathogenesis. SET domain-containing lysine methyltransferase 7 (SETD7) has emerged as one of the key regulators of inflammation. Yet, the function of SETD7 in regulating inflammation-induced ASM cell proliferation and invasion remains unclear. In the present study, we aimed to investigate the function of SETD7 in regulating ASM cell proliferation and invasion induced by tumor necrosis factor (TNF)-α in vitro. Our results showed that SETD7 expression was upregulated in ASM cells stimulated with TNF-α. Silencing SETD7 significantly decreased TNF-α-induced ASM cell proliferation and migration, while SETD7 overexpression exhibited the opposite effect. Notably, silencing SETD7 decreased the activation of nuclear factor (NF)-κB and reduced the expression of CD38 induced by TNF-α. Blocking NF-κB activation significantly abrogated the promotional effect of SETD7 overexpression on CD38 expression. Moreover, overexpression of CD38 partially reversed the inhibitory effect of SETD7 silencing on TNF-α-induced ASM cell proliferation and migration. Overall, these results demonstrate that SETD7 regulates TNF-α-induced ASM cell proliferation and migration through modulation of NF-κB/CD38 signaling, suggesting a potential role of SETD7 in asthma airway remodeling.
Collapse
|
20
|
Siveen KS, Prabhu KS, Parray AS, Merhi M, Arredouani A, Chikri M, Uddin S, Dermime S, Mohammad RM, Steinhoff M, Janahi IA, Azizi F. Evaluation of cationic channel TRPV2 as a novel biomarker and therapeutic target in Leukemia-Implications concerning the resolution of pulmonary inflammation. Sci Rep 2019; 9:1554. [PMID: 30733502 PMCID: PMC6367460 DOI: 10.1038/s41598-018-37469-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/06/2018] [Indexed: 12/20/2022] Open
Abstract
Patients treated during leukemia face the risk of complications including pulmonary dysfunction that may result from infiltration of leukemic blast cells (LBCs) into lung parenchyma and interstitium. In LBCs, we demonstrated that transient receptor potential vanilloid type 2 channel (TRPV2), reputed for its role in inflammatory processes, exhibited oncogenic activity associated with alteration of its molecular expression profile. TRPV2 was overexpressed in LBCs compared to normal human peripheral blood mononuclear cells (PBMCs). Additionally, functional full length isoform and nonfunctional short form pore-less variant of TRPV2 protein were up-regulated and down-regulated respectively in LBCs. However, the opposite was found in PBMCs. TRPV2 silencing or pharmacological targeting by Tranilast (TL) or SKF96365 (SKF) triggered caspace-mediated apoptosis and cell cycle arrest. TL and SKF inhibited chemotactic peptide fMLP-induced response linked to TRPV2 Ca2+ activity, and down-regulated expression of surface marker CD38 involved in leukemia and lung airway inflammation. Challenging lung airway epithelial cells (AECs) with LBCs decreased (by more than 50%) transepithelial resistance (TER) denoting barrier function alteration. Importantly, TL prevented such loss in TER. Therefore, TRPV2 merits further exploration as a pharmacodynamic biomarker for leukemia patients (with pulmonary inflammation) who might be suitable for a novel [adjuvant] therapeutic strategy based on TL.
Collapse
Affiliation(s)
- Kodappully S Siveen
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Aeijaz S Parray
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- National Center for Cancer Care and Research-Hamad Medical Corporation, Doha, Qatar
| | | | - Mohamed Chikri
- Qatar Biomedical Research Institute, Qatar Foundation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- National Center for Cancer Care and Research-Hamad Medical Corporation, Doha, Qatar
| | - Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Fouad Azizi
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
21
|
Role of microRNA in severe asthma. Respir Investig 2018; 57:9-19. [PMID: 30455067 DOI: 10.1016/j.resinv.2018.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 12/23/2022]
Abstract
The various roles of microRNAs (miRNAs) in the epigenetic regulation of human disease are gaining importance as areas of research, and a better understanding of these roles may identify targets for development of novel therapies for severe asthma. MiRNAs, a class of small non-coding RNAs that serve as post-transcriptional gene repressors, are recognized as critical components in regulating tissue homeostasis. Alteration in miRNA expression disrupts homeostasis and is an underlying mechanism for development of chronic respiratory diseases, including asthma. Differential profiles of miRNA expression are involved in inflammation and remodeling pathogenicity via activating airway structural cells and immune cells and inducing cytokine releases. miRNA action leads to asthma progression from mild to severe stages. Here, current knowledge of the heterogeneous roles of miRNAs in severe asthma, including biological mechanisms underlying Th2 and macrophage polarization, type 2 innate lymphoid cell (ILC2) biology regulation, steroid-resistant asthma phenotype, airway smooth muscle (ASM) dysfunction, and impaired anti-viral innate immune, are reviewed.
Collapse
|
22
|
Yuan X, Bhat OM, Meng N, Lohner H, Li PL. Protective Role of Autophagy in Nlrp3 Inflammasome Activation and Medial Thickening of Mouse Coronary Arteries. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2948-2959. [PMID: 30273598 DOI: 10.1016/j.ajpath.2018.08.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 07/16/2018] [Accepted: 08/28/2018] [Indexed: 12/13/2022]
Abstract
We hypothesized that autophagy and associated lysosome function serve as a critical modulator during Nod-like receptor family pyrin domain containing 3 (Nlrp3) inflammasome activation on proatherogenic stimuli. We first demonstrated that 7-ketocholesterol stimulated Nlrp3 inflammasome formation and activation as shown by increased colocalization of inflammasome components [Nlrp3 versus apoptosis associated speck-like protein (Asc) or caspase-1] and enhanced cleavage of caspase-1 into active caspase-1 to generate IL-1β in coronary artery smooth muscle cells. Deletion of the CD38 gene (CD38-/-) that regulates lysosome function and autophagic flux also led to Nlrp3 inflammasome formation and activation. In the presence of rapamycin, the effects of either 7-ketocholesterol treatment or CD38 gene deletion were abolished. The autophagy inhibitor spautin-1 and the lysosome function blocker bafilomycin A1 also enhanced Nlrp3 inflammasome formation and activation. In animal experiments, we found that increased colocalization of Nlrp3 versus Asc or caspase-1 enhanced IL-1β accumulation and caspase-1 activity in the coronary arterial wall of CD38-/- mice on the Western diet compared with CD38+/+ mice. This increased colocalization was blocked by treatment with rapamycin but enhanced by chloroquine, a water-soluble blocker of autophagic flux. Morphologic examinations confirmed that the media of coronary arteries was significantly thicker in CD38-/- mice on the Western diet than CD38+/+ mice. In conclusion, the deficiency of autophagic flux promotes Nlrp3 inflammasome formation and activation in coronary artery smooth muscle cells on proatherogenic stimulation, leading to medial thickening of the coronary arterial wall.
Collapse
Affiliation(s)
- Xinxu Yuan
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Owais M Bhat
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Nan Meng
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Hannah Lohner
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia.
| |
Collapse
|
23
|
MicroRNA-20b-5p inhibits platelet-derived growth factor-induced proliferation of human fetal airway smooth muscle cells by targeting signal transducer and activator of transcription 3. Biomed Pharmacother 2018; 102:34-40. [PMID: 29549727 DOI: 10.1016/j.biopha.2018.03.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/28/2018] [Accepted: 03/05/2018] [Indexed: 02/08/2023] Open
Abstract
Pediatric asthma is still a health threat to the pediatric population in recent years. The airway remodeling induced by abnormal airway smooth muscle (ASM) cell proliferation is an important cause of asthma. MicroRNAs (miRNAs) are important regulators of ASM cell proliferation. Numerous studies have reported that miR-20b-5p is a critical regulator for cell proliferation. However, whether miR-20b-5p is involved in regulating ASM cell proliferation remains unknown. In this study, we aimed to investigate the potential role of miR-20b-5p in regulating the proliferation of fetal ASM cell induced by platelet-derived growth factor (PDGF). Here, we showed that miR-20b-5p was significantly decreased in fetal ASM cells treated with PDGF. Biological experiments showed that the overexpression of miR-20b-5p inhibited the proliferation while miR-20b-5p inhibition markedly promoted the proliferation of fetal ASM cells. Bioinformatics analysis and luciferase reporter assay showed that miR-20b-5p directly targeted the 3'-UTR of signal transducer and activator of transcription 3 (STAT3). Further data showed that miR-20b-5p negatively regulated the expression of STAT3 in fetal ASM cells. Moreover, miR-20b-5p regulates the transcriptional activity of STAT3 in fetal ASM cells. Overexpression of STAT3 reversed the inhibitory effect of miR-20b-5p overexpression on fetal ASM cell proliferation while the knockdown of STAT3 abrogated the promoted effect of miR-20b-5p inhibition on fetal ASM cell proliferation. Overall, our results show that miR-20b-5p impedes PDGF-induced proliferation of fetal ASM cells through targeting STAT3. Our study suggests that miR-20b-5p may play an important role in airway remodeling during asthma and suggests that miR-20b-5p may serve as a potential therapeutic target for pediatric asthma.
Collapse
|
24
|
Chong L, Zhang W, Yu G, Zhang H, Zhu L, Li H, Shao Y, Li C. High-fat-diet induces airway hyperresponsiveness partly through activating CD38 signaling pathway. Int Immunopharmacol 2018; 56:197-204. [PMID: 29414651 DOI: 10.1016/j.intimp.2018.01.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 01/05/2023]
Abstract
CD38 is a plasma membrane bound multifunctional enzyme. It can be activated by inflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-13, inducing calcium responses to agonist in airway smooth muscle cells (ASMC). Previous studies have found that high-fat-diet (HFD) induced obesity exhibited innate airway hyperresponsiveness (AHR). This study aimed to detect the effect of CD38 signaling pathway on the AHR of overweight/obese mice. The HFD-fed mice exhibited a significantly higher baseline airway resistance (Rn), and the increasing rates of Rn responded to increasing doses of methacholine compared with the LFD-fed mice. High-fat-diet increased CD38 expressions both in lung tissues and primary cultured ASMCs. Besides, preincubation with TNF-α led to a higher expression of CD38 protein and increased intracellular calcium in ASMC of the HFD-fed mice. Furthermore, CD38 gene knockdown through transfection of CD38 siRNA decreased the concentration of intracellular calcium. Additionally, the upregulations of CD38 protein and CD38 mRNA were also found in the lung tissues of HFD-fed mice challenged by ovalbumin (OVA). Collectively, our findings demonstrated a role of CD38 signaling pathway on the AHR of obesity and might be a potential therapeutic target for treating difficult-to-control obese asthma phenotype.
Collapse
Affiliation(s)
- Lei Chong
- Institute of Pediatrics, National Key Clinical Specialty of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weixi Zhang
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gang Yu
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hailin Zhang
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lili Zhu
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haiyan Li
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Youyou Shao
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Changchong Li
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
25
|
Ji Y, Yang X, Su H. Overexpression of microRNA-375 impedes platelet-derived growth factor-induced proliferation and migration of human fetal airway smooth muscle cells by targeting Janus kinase 2. Biomed Pharmacother 2017; 98:69-75. [PMID: 29245068 DOI: 10.1016/j.biopha.2017.12.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/20/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022] Open
Abstract
The abnormal proliferation and migration of airway smooth muscle (ASM) cells play a critical role in airway remodeling during the development of asthma. MicroRNAs (miRNAs) have emerged as critical regulators of ASM cell proliferation and migration in airway remodeling. In this study, we aimed to investigate the potential role of miR-375 in the regulation of platelet-derived growth factor (PDGF)-induced fetal ASM cell proliferation and migration. Our results showed that miR-375 expression was significantly decreased in fetal ASM cells that were treated with PDGF. Functional data showed that overexpression of miR-375 inhibited the proliferation and migration of fetal ASM cells, whereas inhibition of miR-375 enhanced the proliferation and migration of fetal ASM cells. The results of bioinformatics analysis and a dual-luciferase reporter assay showed that miR-375 binds directly to the 3'-untranslated region of Janus kinase 2 (JAK2). Further data confirmed that miR-375 negatively regulates the expression of JAK2 in fetal ASM cells. Moreover, miR-375 also impeded the PDGF-induced activation of signal transducer and activator of transcription 3 (STAT3) in fetal ASM cells. However, restoration of JAK2 expression partially reversed the inhibitory effect of miR-375 on fetal ASM cell proliferation and migration. Overall, our results demonstrate that miR-375 inhibits fetal ASM cell proliferation and migration by targeting JAK2/STAT3 signaling. Our study provides a potential therapeutic target for the development of novel treatment strategies for pediatric asthma.
Collapse
Affiliation(s)
- Yamei Ji
- Department of Paediatrics, Yulin Xingyuan Hospital, Yulin, Shaanxi 719000, China
| | - Xin Yang
- Second Department of Paediatric Internal Medicine, Yulin Children's Hospital, Yulin, Shaanxi 719000, China
| | - Huixia Su
- Second Department of Paediatric Internal Medicine, Yulin Children's Hospital, Yulin, Shaanxi 719000, China.
| |
Collapse
|
26
|
Lee HC, Mark TM, Shah JJ. Practical Approaches to the Management of Dual Refractory Multiple Myeloma. Curr Hematol Malig Rep 2017; 11:148-55. [PMID: 26898556 DOI: 10.1007/s11899-016-0312-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The outcome of myeloma patients' dual refractory to lenalidomide and bortezomib is generally poor and represents a significant clinical challenge with a clear need for new therapeutic approaches. This has prompted the development of next-generation proteasome inhibitors and immunodulatory drugs (IMiDs), as well as new classes of drugs with novel mechanisms of action. As a result, several of these agents have received regulatory approval that have shown promising activity in the dual refractory setting including the second-generation proteasome inhibitor carfilzomib and third-generation IMiD pomalidomide. Moreover, the regulatory approval of several first-in-class drugs for myeloma such as the histone deacetylase (HDAC) inhibitor panobinostat and the anti-CD38 monoclonal antibody daratumumab has further broadened the therapeutic landscape for these patients. Collectively, these advances have provided new treatment strategies in dual refractory myeloma as well as important insights for the development of future studies with rationally designed drug combinations to target this challenging patient population.
Collapse
Affiliation(s)
- Hans C Lee
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 429, Houston, TX, 77030, USA
| | - Tomer M Mark
- Multiple Myeloma Center, Weil Cornell Medical College, 428 E. 72nd street, Suite 300, New York, NY, 10021, USA
| | - Jatin J Shah
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 429, Houston, TX, 77030, USA.
| |
Collapse
|
27
|
Jónsdóttir U, Lang JE. How does autism spectrum disorder affect the risk and severity of childhood asthma? Ann Allergy Asthma Immunol 2017; 118:570-576. [PMID: 28477788 DOI: 10.1016/j.anai.2017.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/16/2017] [Accepted: 02/20/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) and asthma are among the most common chronic disorders in childhood. Both are associated with altered immune regulation and share several risk factors. The effects of ASD on risk for later asthma and asthma severity remain unclear. OBJECTIVE To determine whether ASD in children increases the risk of incident asthma and worsens asthma severity. METHODS We performed 2 distinct analytic designs (case-control and retrospective longitudinal cohort) using a multistate electronic health records database to assess the odds of new asthma and asthma severity among children with ASD. In both designs, children with ASD were matched with children without ASD according to sex, age, race, ethnicity, location, and insurance status. Pulmonary function, controller medication prescriptions, asthma exacerbations, and asthma-related hospitalizations were collected. The effects of ASD on asthma risk and severity were assessed using multivariable linear and logistic regression. RESULTS Among children with asthma, ASD was associated with reduced exacerbations (odds ratio [OR], 0.71; 95% confidence interval [CI], 0.54-0.92), better forced expiratory volume in 1 second/forced vital capacity ratio (0.876 vs 0.841, P < .001), and lower odds of airflow obstruction (OR, 0.53; 95% CI, 0.31-0.90) but had higher odds of asthma controller prescription (OR, 2.18; 95% CI, 1.62-2.93). In a longitudinal analysis of children without asthma, ASD was found to be protective for new asthma (OR, 0.44; 95% CI, 0.26-0.74). CONCLUSION Among children with asthma, concomitant ASD is associated with better asthma-related outcomes but a higher controller treatment burden. In addition, our data did not support ASD as a risk factor for incident asthma.
Collapse
Affiliation(s)
| | - Jason E Lang
- Division of Pulmonary and Sleep Medicine, Nemours Children's Hospital, Orlando, Florida; Division of Allergy/Immunology and Pulmonary Medicine, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
28
|
Villegas-Ospina S, Aguilar-Jimenez W, Gonzalez SM, Rugeles MT. Vitamin D modulates the expression of HLA-DR and CD38 after in vitro activation of T-cells. Horm Mol Biol Clin Investig 2017; 29:93-103. [PMID: 28222027 DOI: 10.1515/hmbci-2016-0037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/29/2016] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Vitamin D (VitD) is an anti-inflammatory hormone; however, some evidence shows that VitD may induce the expression of activation markers, such as CD38 and HLA-DR. We explored its effect on the expression of these markers on CD4+ and CD8+ T-cells in vitro, and their potential correlations in vivo. MATERIALS AND METHODS CD38 and HLA-DR expression was measured by flow cytometry in PHA/IL-2-activated mononuclear cells cultured under VitD precursors: three cholecalciferol (10-11M, 10-9M, 10-7M; n=11) and two calcidiol (40 ng/mL, 80 ng/mL; n=9) concentrations. The correlation between the expression of these markers in freshly isolated blood cells and serum levels of calcidiol was also explored (n=10). RESULTS Cholecalciferol at 10-7M increased the proportion of CD4+ CD38+ and CD8+ CD38+ cells, and decreased CD8+HLA-DR+ cells. As co-expression, it increased the CD38+HLA-DR- and decreased CD38-HLA-DR+ subpopulations in both CD4+ and CD8+ T-cells, and decreased CD4+CD38-HLA-DR- and CD8+ CD38+HLA-DR+; whereas both calcidiol concentrations decreased the proliferation of CD38-HLA-DR- and CD38-HLA-DR+ subpopulations. Both forms of VitD increased the number of CD38 molecules per cell. In contrast, there was a positive but non-significant correlation between serum calcidiol levels and the expression of CD38 and HLA-DR in CD4+ and CD8+ T-cells. CONCLUSION Although no significant correlations were observed in vivo in healthy subjects, VitD treatment in vitro modulated immune activation by increasing the expression of CD38 and decreasing the proliferation of HLA-DR+ and resting cells, which may correlate with improved effector and decreased proliferative capabilities. These results highlight the potential use of VitD as therapeutic strategy in immune disorders.
Collapse
|
29
|
Peng QY, Zou Y, Zhang LN, Ai ML, Liu W, Ai YH. Blocking Cyclic Adenosine Diphosphate Ribose-mediated Calcium Overload Attenuates Sepsis-induced Acute Lung Injury in Rats. Chin Med J (Engl) 2017; 129:1725-30. [PMID: 27411462 PMCID: PMC4960964 DOI: 10.4103/0366-6999.185854] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background: Acute lung injury (ALI) is a common complication of sepsis that is associated with high mortality. Intracellular Ca2+ overload plays an important role in the pathophysiology of sepsis-induced ALI, and cyclic adenosine diphosphate ribose (cADPR) is an important regulator of intracellular Ca2+ mobilization. The cluster of differentiation 38 (CD38)/cADPR pathway has been found to play roles in multiple inflammatory processes but its role in sepsis-induced ALI is still unknown. This study aimed to investigate whether the CD38/cADPR signaling pathway is activated in sepsis-induced ALI and whether blocking cADPR-mediated calcium overload attenuates ALI. Methods: Septic rat models were established by cecal ligation and puncture (CLP). Rats were divided into the sham group, the CLP group, and the CLP+ 8-bromo-cyclic adenosine diphosphate ribose (8-Br-cADPR) group. Nicotinamide adenine dinucleotide (NAD+), cADPR, CD38, and intracellular Ca2+ levels in the lung tissues were measured at 6, 12, 24, and 48 h after CLP surgery. Lung histologic injury, tumor necrosis factor (TNF)-α, malondialdehyde (MDA) levels, and superoxide dismutase (SOD) activities were measured. Results: NAD+, cADPR, CD38, and intracellular Ca2+ levels in the lungs of septic rats increased significantly at 24 h after CLP surgery. Treatment with 8-Br-cADPR, a specific inhibitor of cADPR, significantly reduced intracellular Ca2+ levels (P = 0.007), attenuated lung histological injury (P = 0.023), reduced TNF-α and MDA levels (P < 0.001 and P = 0.002, respectively) and recovered SOD activity (P = 0.031) in the lungs of septic rats. Conclusions: The CD38/cADPR pathway is activated in the lungs of septic rats, and blocking cADPR-mediated calcium overload with 8-Br-cADPR protects against sepsis-induced ALI.
Collapse
Affiliation(s)
- Qian-Yi Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yu Zou
- Department of Anesthesia, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Li-Na Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Mei-Lin Ai
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wei Liu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yu-Hang Ai
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
30
|
Deshpande DA, Guedes AGP, Lund FE, Subramanian S, Walseth TF, Kannan MS. CD38 in the pathogenesis of allergic airway disease: Potential therapeutic targets. Pharmacol Ther 2016; 172:116-126. [PMID: 27939939 DOI: 10.1016/j.pharmthera.2016.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CD38 is an ectoenzyme that catalyzes the conversion of β-nicotinamide adenine dinucleotide (β-NAD) to cyclic adenosine diphosphoribose (cADPR) and adenosine diphosphoribose (ADPR) and NADP to nicotinic acid adenine dinucleotide phosphate (NAADP) and adenosine diphosphoribose-2'-phosphate (ADPR-P). The metabolites of NAD and NADP have roles in calcium signaling in different cell types including airway smooth muscle (ASM) cells. In ASM cells, inflammatory cytokines augment CD38 expression and to a greater magnitude in cells from asthmatics, indicating a greater capacity for the generation of cADPR and ADPR in ASM from asthmatics. CD38 deficient mice develop attenuated airway responsiveness to inhaled methacholine following allergen sensitization and challenge compared to wild-type mice indicating its potential role in asthma. Regulation of CD38 expression in ASM cells is achieved by mitogen activated protein kinases, specific isoforms of PI3 kinases, the transcription factors NF-κB and AP-1, and post-transcriptionally by microRNAs. This review will focus on the role of CD38 in intracellular calcium regulation in ASM, contribution to airway inflammation and airway hyperresponsiveness in mouse models of allergic airway inflammation, the transcriptional and post-transcriptional mechanisms of regulation of expression, and outline approaches to inhibit its expression and activity.
Collapse
Affiliation(s)
| | - Alonso G P Guedes
- Department of Veterinary Clinical Sciences, University of Minnesota at Twin Cities, USA
| | - Frances E Lund
- Department of Microbiology, University of Alabama at Birmingham, USA
| | | | - Timothy F Walseth
- Department of Pharmacology, University of Minnesota at Twin Cities, USA
| | - Mathur S Kannan
- Department of Veterinary and Biomedical Sciences, University of Minnesota at Twin Cities, USA.
| |
Collapse
|
31
|
Prakash YS. Emerging concepts in smooth muscle contributions to airway structure and function: implications for health and disease. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1113-L1140. [PMID: 27742732 DOI: 10.1152/ajplung.00370.2016] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/06/2016] [Indexed: 12/15/2022] Open
Abstract
Airway structure and function are key aspects of normal lung development, growth, and aging, as well as of lung responses to the environment and the pathophysiology of important diseases such as asthma, chronic obstructive pulmonary disease, and fibrosis. In this regard, the contributions of airway smooth muscle (ASM) are both functional, in the context of airway contractility and relaxation, as well as synthetic, involving production and modulation of extracellular components, modulation of the local immune environment, cellular contribution to airway structure, and, finally, interactions with other airway cell types such as epithelium, fibroblasts, and nerves. These ASM contributions are now found to be critical in airway hyperresponsiveness and remodeling that occur in lung diseases. This review emphasizes established and recent discoveries that underline the central role of ASM and sets the stage for future research toward understanding how ASM plays a central role by being both upstream and downstream in the many interactive processes that determine airway structure and function in health and disease.
Collapse
Affiliation(s)
- Y S Prakash
- Departments of Anesthesiology, and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
32
|
Liu L, Pan Y, Song Y, Su X, Ke R, Yang L, Gao L, Li M. Activation of AMPK α2 inhibits airway smooth muscle cells proliferation. Eur J Pharmacol 2016; 791:235-243. [PMID: 27600020 DOI: 10.1016/j.ejphar.2016.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 09/02/2016] [Accepted: 09/02/2016] [Indexed: 12/31/2022]
Abstract
The aims of the present study were to examine the effect of adenosine monophosphate-activated protein kinase (AMPK) activation on airway smooth muscle cells (ASMCs) proliferation and to address its potential mechanisms. Platelet derived growth factor (PDGF) activated phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway, and this in turn up-regulated S-phase kinase-associated protein 2 (Skp2) and consequently reduced cyclin dependent kinase inhibitor 1B (p27) leading to ASMCs proliferation. Pre-incubation of cells with metformin, an AMPK activator, blocked PDGF-induced activation of mTOR and its downstream targets changes of Skp2 and p27 without changing Akt phosphorylation and inhibited ASMCs proliferation. Transfection of ASMCs with AMPK α2-specific small interfering RNA (siRNA) reversed the effect of metformin on mTOR phosphorylation, Skp2 and p27 protein expression and cell proliferation. Our study suggests that activation of AMPK, particularly AMPK α2, negatively regulates mTOR activity to suppress ASMCs proliferation and therefore has a potential value in the prevention and treatment of asthma by negatively modulating airway remodeling.
Collapse
Affiliation(s)
- Lu Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| | - Yilin Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| | - Yang Song
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| | - Xiaofan Su
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| | - Rui Ke
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| | - Lan Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| | - Li Gao
- Division of Allergy and Clinical Immunology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA.
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
33
|
Aravamudan B, Thompson MA, Pabelick CM, Prakash YS. Mechanisms of BDNF regulation in asthmatic airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2016; 311:L270-9. [PMID: 27317689 DOI: 10.1152/ajplung.00414.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 06/09/2016] [Indexed: 12/17/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a neurotrophin produced by airway smooth muscle (ASM), enhances inflammation effects on airway contractility, supporting the idea that locally produced growth factors influence airway diseases such as asthma. We endeavored to dissect intrinsic mechanisms regulating endogenous, as well as inflammation (TNF-α)-induced BDNF secretion in ASM of nonasthmatic vs. asthmatic humans. We focused on specific Ca(2+) regulation- and inflammation-related signaling cascades and quantified BDNF secretion. We find that TNF-α enhances BDNF release by ASM cells, via several mechanisms relevant to asthma, including transient receptor potential channels TRPC3 and TRPC6 (but not TRPC1), ERK 1/2, PI3K, PLC, and PKC cascades, Rho kinase, and transcription factors cAMP response element binding protein and nuclear factor of activated T cells. Basal BDNF expression and secretion are elevated in asthmatic ASM and increase further with TNF-α exposure, involving many of these regulatory mechanisms. We conclude that airway BDNF secretion is regulated at multiple levels, providing a basis for autocrine effects of BDNF under conditions of inflammation and disease, with potential downstream influences on contractility and remodeling.
Collapse
Affiliation(s)
| | | | - Christina M Pabelick
- Departments of Anesthesiology, Mayo Clinic, Rochester, Minnesota; and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y S Prakash
- Departments of Anesthesiology, Mayo Clinic, Rochester, Minnesota; and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
34
|
Dileepan M, Sarver AE, Rao SP, Panettieri RA, Subramanian S, Kannan MS. MicroRNA Mediated Chemokine Responses in Human Airway Smooth Muscle Cells. PLoS One 2016; 11:e0150842. [PMID: 26998837 PMCID: PMC4801396 DOI: 10.1371/journal.pone.0150842] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/19/2016] [Indexed: 01/25/2023] Open
Abstract
Airway smooth muscle (ASM) cells play a critical role in the pathophysiology of asthma due to their hypercontractility and their ability to proliferate and secrete inflammatory mediators. microRNAs (miRNAs) are gene regulators that control many signaling pathways and thus serve as potential therapeutic alternatives for many diseases. We have previously shown that miR-708 and miR-140-3p regulate the MAPK and PI3K signaling pathways in human ASM (HASM) cells following TNF-α exposure. In this study, we investigated the regulatory effect of these miRNAs on other asthma-related genes. Microarray analysis using the Illumina platform was performed with total RNA extracted from miR-708 (or control miR)-transfected HASM cells. Inhibition of candidate inflammation-associated gene expression was further validated by qPCR and ELISA. The most significant biologic functions for the differentially expressed gene set included decreased inflammatory response, cytokine expression and signaling. qPCR revealed inhibition of expression of CCL11, CXCL10, CCL2 and CXCL8, while the release of CCL11 was inhibited in miR-708-transfected cells. Transfection of cells with miR-140-3p resulted in inhibition of expression of CCL11, CXCL12, CXCL10, CCL5 and CXCL8 and of TNF-α-induced CXCL12 release. In addition, expression of RARRES2, CD44 and ADAM33, genes known to contribute to the pathophysiology of asthma, were found to be inhibited in miR-708-transfected cells. These results demonstrate that miR-708 and miR-140-3p exert distinct effects on inflammation-associated gene expression and biological function of ASM cells. Targeting these miRNA networks may provide a novel therapeutic mechanism to down-regulate airway inflammation and ASM proliferation in asthma.
Collapse
Affiliation(s)
- Mythili Dileepan
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Anne E. Sarver
- Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Savita P. Rao
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Reynold A. Panettieri
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Subbaya Subramanian
- Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Mathur S. Kannan
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
35
|
Kaphalia L, Kalita M, Kaphalia BS, Calhoun WJ. Effects of acute ethanol exposure on cytokine production by primary airway smooth muscle cells. Toxicol Appl Pharmacol 2015; 292:85-93. [PMID: 26721307 DOI: 10.1016/j.taap.2015.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 12/08/2015] [Accepted: 12/17/2015] [Indexed: 01/17/2023]
Abstract
Both chronic and binge alcohol abuse can be significant risk factors for inflammatory lung diseases such as acute respiratory distress syndrome and chronic obstructive pulmonary disease. However, metabolic basis of alcohol-related lung disease is not well defined, and may include key metabolites of ethanol [EtOH] in addition to EtOH itself. Therefore, we investigated the effects of EtOH, acetaldehyde [ACE], and fatty acid ethyl esters [FAEEs] on oxidative stress, endoplasmic reticulum (ER) stress, AMP-activated protein kinase (AMPK) signaling and nuclear translocation of phosphorylated (p)-NF-κB p65 in primary human airway smooth muscle (HASM) cells stimulated to produce cytokines using LPS exposure. Both FAEEs and ACE induced evidence of cellular oxidative stress and ER stress, and increased p-NF-κB in nuclear extracts. EtOH and its metabolites decreased p-AMPKα activation, and induced expression of fatty acid synthase, and decreased expression of sirtuin 1. In general, EtOH decreased secretion of IP-10, IL-6, eotaxin, GCSF, and MCP-1. However, FAEEs and ACE increased these cytokines, suggesting that both FAEEs and ACE as compared to EtOH itself are proinflammatory. A direct effect of EtOH could be consistent with blunted immune response. Collectively, these two features of EtOH exposure, coupled with the known inhibition of innate immune response in our model might explain some clinical manifestations of EtOH exposure in the lung.
Collapse
Affiliation(s)
- Lata Kaphalia
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Mridul Kalita
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Bhupendra S Kaphalia
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - William J Calhoun
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
36
|
Deshpande DA, Dileepan M, Walseth TF, Subramanian S, Kannan MS. MicroRNA Regulation of Airway Inflammation and Airway Smooth Muscle Function: Relevance to Asthma. Drug Dev Res 2015; 76:286-95. [PMID: 26587803 DOI: 10.1002/ddr.21267] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Genetic and environmental factors contribute to the onset and severity of asthma. Molecular pathogenesis of asthma involves changes in gene expression by a variety of inflammatory mediators acting in autocrine and paracrine fashion on effector cells of the airways. Transcriptional regulation of gene expression in resident airway cells has been studied extensively. However, protein function in a target cell can be regulated at multiple levels starting from transcription followed by post-transcription, translation, and post-translation steps. In this context, small noncoding RNAs known as microRNAs (miRNAs) have evolved as one of the key regulators of gene expression post-transcriptionally. Most importantly, miRNA expression is dynamic in nature and can be regulated by a variety of external stimuli. Altered expression of individual or a group of miRNAs is thought to contribute to human diseases. Recent studies have implicated differential expression of miRNAs in the lungs during inflammation. Most importantly, advanced biochemical and molecular tools could be used to manipulate miRNA expression thereby effecting functional changes in target cells and organ systems. This review summarizes the current understanding of miRNA in the regulation of airway function in health and disease, and highlights the potential clinical utility of mRNAs as biomarkers of airway diseases and as potential therapeutic targets.
Collapse
Affiliation(s)
- D A Deshpande
- Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - M Dileepan
- Department of Veterinary and Biomedical Sciences, University of Minnesota, MN, USA
| | - T F Walseth
- Department of Pharmacology, University of Minnesota, MN, USA
| | - S Subramanian
- Department of Surgery, University of Minnesota, MN, USA
| | - M S Kannan
- Department of Veterinary and Biomedical Sciences, University of Minnesota, MN, USA
| |
Collapse
|
37
|
Comhair SAA, McDunn J, Bennett C, Fettig J, Erzurum SC, Kalhan SC. Metabolomic Endotype of Asthma. THE JOURNAL OF IMMUNOLOGY 2015; 195:643-50. [PMID: 26048149 DOI: 10.4049/jimmunol.1500736] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/13/2015] [Indexed: 11/19/2022]
Abstract
Metabolomics, the quantification of small biochemicals in plasma and tissues, can provide insight into complex biochemical processes and enable the identification of biomarkers that may serve as therapeutic targets. We hypothesized that the plasma metabolome of asthma would reveal metabolic consequences of the specific immune and inflammatory responses unique to endotypes of asthma. The plasma metabolomic profiles of 20 asthmatic subjects and 10 healthy controls were examined using an untargeted global and focused metabolomic analysis. Individuals were classified based on clinical definitions of asthma severity or by levels of fraction of exhaled NO (FENO), a biomarker of airway inflammation. Of the 293 biochemicals identified in the plasma, 25 were significantly different among asthma and healthy controls (p < 0.05). Plasma levels of taurine, lathosterol, bile acids (taurocholate and glycodeoxycholate), nicotinamide, and adenosine-5-phosphate were significantly higher in asthmatics compared with healthy controls. Severe asthmatics had biochemical changes related to steroid and amino acid/protein metabolism. Asthmatics with high FENO, compared with those with low FENO, had higher levels of plasma branched-chain amino acids and bile acids. Asthmatics have a unique plasma metabolome that distinguishes them from healthy controls and points to activation of inflammatory and immune pathways. The severe asthmatic and high FENO asthmatic have unique endotypes that suggest changes in NO-associated taurine transport and bile acid metabolism.
Collapse
Affiliation(s)
- Suzy A A Comhair
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; Cleveland Clinic, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| | | | - Carole Bennett
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jade Fettig
- Cleveland Clinic, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| | - Serpil C Erzurum
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; Cleveland Clinic, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195; Respiratory Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Satish C Kalhan
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; Cleveland Clinic, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195;
| |
Collapse
|