1
|
He Y, Han Y, Zou L, Yao T, Zhang Y, Lv X, Jiang M, Long L, Li M, Cheng X, Jiang G, Peng Z, Tao L, Meng J, Xie W. Succinate promotes pulmonary fibrosis through GPR91 and predicts death in idiopathic pulmonary fibrosis. Sci Rep 2024; 14:14376. [PMID: 38909094 PMCID: PMC11193722 DOI: 10.1038/s41598-024-64844-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/13/2024] [Indexed: 06/24/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is believed to be associated with a notable disruption of cellular energy metabolism. By detecting the changes of energy metabolites in the serum of patients with pulmonary fibrosis, we aimed to investigate the diagnostic and prognostic value of energy metabolites in IPF, and further elucidated the mechanism of their involvement in pulmonary fibrosis. Through metabolomics research, it was discovered that the TCA cycle intermediates changed dramatically in IPF patients. In another validation cohort of 55 patients with IPF compared to 19 healthy controls, it was found that succinate, an intermediate product of TCA cycle, has diagnostic and prognostic value in IPF. The cut-off levels of serum succinate were 98.36 μM for distinguishing IPF from healthy controls (sensitivity, 83.64%; specificity, 63.16%; likelihood ratio, 2.27, respectively). Moreover, a high serum succinate level was independently associated with higher rates of disease progression (OR 13.087, 95%CI (2.819-60.761)) and mortality (HR 3.418, 95% CI (1.308-8.927)). In addition, accumulation of succinate and increased expression of the succinate receptor GPR91 were found in both IPF patients and BLM mouse models of pulmonary fibrosis. Reducing succinate accumulation in BLM mice alleviated pulmonary fibrosis and 21d mortality, while exogenous administration of succinate can aggravate pulmonary fibrosis in BLM mice. Furthermore, GPR91 deficiency protected against lung fibrosis caused by BLM. In vitro, succinate promoted the activation of lung fibroblasts by activating ERK pathway through GPR91. In summary, succinate is a promising biomarker for diagnosis and prognosis of IPF. The accumulation of succinate may promote fibroblast activation through GPR91 and pulmonary fibrosis.
Collapse
Affiliation(s)
- Yijun He
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Yuanyuan Han
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Lijun Zou
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Tingting Yao
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Yan Zhang
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Xin Lv
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Mao Jiang
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Lingzhi Long
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Mengyu Li
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Xiaoyun Cheng
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Department of Pulmonary and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Guoliang Jiang
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Zhangzhe Peng
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| | - Lijian Tao
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| | - Jie Meng
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan, China.
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China.
- National International Collaborative Research Center for Medical Metabolomics, Changsha, China.
| | - Wei Xie
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
2
|
Fioretto BS, Rosa I, Tani A, Andreucci E, Romano E, Sgambati E, Manetti M. Blockade of Sialylation with Decrease in Polysialic Acid Levels Counteracts Transforming Growth Factor β1-Induced Skin Fibroblast-to-Myofibroblast Transition. Cells 2024; 13:1067. [PMID: 38920695 PMCID: PMC11201575 DOI: 10.3390/cells13121067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Aberrant sialylation with overexpression of the homopolymeric glycan polysialic acid (polySia) was recently reported in fibroblasts from fibrotic skin lesions. Yet, whether such a rise in polySia levels or sialylation in general may be functionally implicated in profibrotic activation of fibroblasts and their transition to myofibroblasts remains unknown. Therefore, we herein explored whether inhibition of sialylation could interfere with the process of skin fibroblast-to-myofibroblast transition induced by the master profibrotic mediator transforming growth factor β1 (TGFβ1). Adult human skin fibroblasts were pretreated with the competitive pan-sialyltransferase inhibitor 3-Fax-peracetyl-Neu5Ac (3-Fax) before stimulation with recombinant human TGFβ1, and then analyzed for polySia expression, cell viability, proliferation, migratory ability, and acquisition of myofibroblast-like morphofunctional features. Skin fibroblast stimulation with TGFβ1 resulted in overexpression of polySia, which was effectively blunted by 3-Fax pre-administration. Pretreatment with 3-Fax efficiently lessened TGFβ1-induced skin fibroblast proliferation, migration, changes in cell morphology, and phenotypic and functional differentiation into myofibroblasts, as testified by a significant reduction in FAP, ACTA2, COL1A1, COL1A2, and FN1 gene expression, and α-smooth muscle actin, N-cadherin, COL1A1, and FN-EDA protein levels, as well as a reduced contractile capability. Moreover, skin fibroblasts pre-administered with 3-Fax displayed a significant decrease in Smad3-dependent canonical TGFβ1 signaling. Collectively, our in vitro findings demonstrate for the first time that aberrant sialylation with increased polySia levels has a functional role in skin fibroblast-to-myofibroblast transition and suggest that competitive sialyltransferase inhibition might offer new therapeutic opportunities against skin fibrosis.
Collapse
Affiliation(s)
- Bianca Saveria Fioretto
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (B.S.F.); (I.R.); (A.T.)
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (B.S.F.); (I.R.); (A.T.)
- Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Alessia Tani
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (B.S.F.); (I.R.); (A.T.)
- Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Elena Andreucci
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy;
| | - Eloisa Romano
- Section of Internal Medicine, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy;
| | - Eleonora Sgambati
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, 86090 Isernia, Italy;
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (B.S.F.); (I.R.); (A.T.)
- Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| |
Collapse
|
3
|
Koziol-White C, Gebski E, Cao G, Panettieri RA. Precision cut lung slices: an integrated ex vivo model for studying lung physiology, pharmacology, disease pathogenesis and drug discovery. Respir Res 2024; 25:231. [PMID: 38824592 PMCID: PMC11144351 DOI: 10.1186/s12931-024-02855-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/18/2024] [Indexed: 06/03/2024] Open
Abstract
Precision Cut Lung Slices (PCLS) have emerged as a sophisticated and physiologically relevant ex vivo model for studying the intricacies of lung diseases, including fibrosis, injury, repair, and host defense mechanisms. This innovative methodology presents a unique opportunity to bridge the gap between traditional in vitro cell cultures and in vivo animal models, offering researchers a more accurate representation of the intricate microenvironment of the lung. PCLS require the precise sectioning of lung tissue to maintain its structural and functional integrity. These thin slices serve as invaluable tools for various research endeavors, particularly in the realm of airway diseases. By providing a controlled microenvironment, precision-cut lung slices empower researchers to dissect and comprehend the multifaceted interactions and responses within lung tissue, thereby advancing our understanding of pulmonary pathophysiology.
Collapse
Affiliation(s)
- Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA.
| | - Eric Gebski
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Gaoyaun Cao
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| |
Collapse
|
4
|
Mohanan A, Washimkar KR, Mugale MN. Unraveling the interplay between vital organelle stress and oxidative stress in idiopathic pulmonary fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119676. [PMID: 38242330 DOI: 10.1016/j.bbamcr.2024.119676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/22/2023] [Accepted: 01/10/2024] [Indexed: 01/21/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease characterized by excessive accumulation of extracellular matrix, leading to irreversible fibrosis. Emerging evidence suggests that endoplasmic reticulum (ER) stress, mitochondrial stress, and oxidative stress pathways play crucial roles in the pathogenesis of IPF. ER stress occurs when the protein folding capacity of the ER is overwhelmed, triggering the unfolded protein response (UPR) and contributing to protein misfolding and cellular stress in IPF. Concurrently, mitochondrial dysfunction involving dysregulation of key regulators, including PTEN-induced putative kinase 1 (PINK1), Parkin, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and sirtuin 3 (SIRT3), disrupts mitochondrial homeostasis and impairs cellular energy metabolism. This leads to increased reactive oxygen species (ROS) production, release of pro-fibrotic mediators, and activation of fibrotic pathways, exacerbating IPF progression. The UPR-induced ER stress further disrupts mitochondrial metabolism, resulting in altered mitochondrial mechanisms that increase the generation of ROS, resulting in further ER stress, creating a feedback loop that contributes to the progression of IPF. Oxidative stress also plays a pivotal role in IPF, as ROS-mediated activation of TGF-β, NF-κB, and MAPK pathways promotes inflammation and fibrotic responses. This review mainly focuses on the links between ER stress, mitochondrial dysfunctions, and oxidative stress with different signaling pathways involved in IPF. Understanding these mechanisms and targeting key molecules within these pathways may offer promising avenues for intervention.
Collapse
Affiliation(s)
- Anushree Mohanan
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India
| | - Kaveri R Washimkar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
5
|
Lettieri S, Bertuccio FR, del Frate L, Perrotta F, Corsico AG, Stella GM. The Plastic Interplay between Lung Regeneration Phenomena and Fibrotic Evolution: Current Challenges and Novel Therapeutic Perspectives. Int J Mol Sci 2023; 25:547. [PMID: 38203718 PMCID: PMC10779349 DOI: 10.3390/ijms25010547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Interstitial lung diseases (ILDs) are a heterogeneous group of pulmonary disorders characterized by variable degrees of inflammation, interstitial thickening, and fibrosis leading to distortion of the pulmonary architecture and gas exchange impairment. Among them, idiopathic pulmonary fibrosis (IPF) displays the worst prognosis. The only therapeutic options consist of the two antifibrotic drugs, pirfenidone and nintedanib, which limit fibrosis progression but do not reverse the lung damage. The shift of the pathogenetic paradigm from inflammatory disease to epithelium-derived disease has definitively established the primary role of type II alveolar cells, which lose their epithelial phenotype and acquire a mesenchymal phenotype with production of collagen and extracellular matrix (EMC) deposition. Some predisposing environmental and genetic factors (e.g., smoke, pollution, gastroesophageal reflux, variants of telomere and surfactant genes) leading to accelerated senescence set a pro-fibrogentic microenvironment and contribute to the loss of regenerative properties of type II epithelial cells in response to pathogenic noxae. This review provides a complete overview of the different pathogenetic mechanisms leading to the development of IPF. Then, we summarize the currently approved therapies and the main clinical trials ongoing. Finally, we explore the potentialities offered by agents not only interfering with the processes of fibrosis but also restoring the physiological properties of alveolar regeneration, with a particular focus on potentialities and concerns about cell therapies based on mesenchymal stem cells (MSCs), whose anti-inflammatory and immunomodulant properties have been exploited in other fibrotic diseases, such as graft versus host disease (GVHD) and COVID-19-related ARDS.
Collapse
Affiliation(s)
- Sara Lettieri
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Francesco R. Bertuccio
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Lucia del Frate
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Fabio Perrotta
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, 80055 Naples, Italy;
| | - Angelo G. Corsico
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giulia M. Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
6
|
Castillejos-López M, Romero Y, Varela-Ordoñez A, Flores-Soto E, Romero-Martinez BS, Velázquez-Cruz R, Vázquez-Pérez JA, Ruiz V, Gomez-Verjan JC, Rivero-Segura NA, Camarena Á, Torres-Soria AK, Gonzalez-Avila G, Sommer B, Solís-Chagoyán H, Jaimez R, Torres-Espíndola LM, Aquino-Gálvez A. Hypoxia Induces Alterations in the Circadian Rhythm in Patients with Chronic Respiratory Diseases. Cells 2023; 12:2724. [PMID: 38067152 PMCID: PMC10706372 DOI: 10.3390/cells12232724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/08/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
The function of the circadian cycle is to determine the natural 24 h biological rhythm, which includes physiological, metabolic, and hormonal changes that occur daily in the body. This cycle is controlled by an internal biological clock that is present in the body's tissues and helps regulate various processes such as sleeping, eating, and others. Interestingly, animal models have provided enough evidence to assume that the alteration in the circadian system leads to the appearance of numerous diseases. Alterations in breathing patterns in lung diseases can modify oxygenation and the circadian cycles; however, the response mechanisms to hypoxia and their relationship with the clock genes are not fully understood. Hypoxia is a condition in which the lack of adequate oxygenation promotes adaptation mechanisms and is related to several genes that regulate the circadian cycles, the latter because hypoxia alters the production of melatonin and brain physiology. Additionally, the lack of oxygen alters the expression of clock genes, leading to an alteration in the regularity and precision of the circadian cycle. In this sense, hypoxia is a hallmark of a wide variety of lung diseases. In the present work, we intended to review the functional repercussions of hypoxia in the presence of asthma, chronic obstructive sleep apnea, lung cancer, idiopathic pulmonary fibrosis, obstructive sleep apnea, influenza, and COVID-19 and its repercussions on the circadian cycles.
Collapse
Affiliation(s)
- Manuel Castillejos-López
- Departamento de Epidemiología e Infectología Hospitalaria, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City 14080, Mexico;
| | - Yair Romero
- Facultad de Ciencias, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico;
| | - Angelica Varela-Ordoñez
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.V.-O.); (A.K.T.-S.)
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico; (E.F.-S.); (B.S.R.-M.); (R.J.)
| | - Bianca S. Romero-Martinez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico; (E.F.-S.); (B.S.R.-M.); (R.J.)
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Joel Armando Vázquez-Pérez
- Laboratorio de Biología Molecular de Enfermedades Emergentes y EPOC, Instituto Nacional de Enferdades Respiratorias Ismael Cosío Villegas (INER), Mexico City 14080, Mexico;
| | - Víctor Ruiz
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City 14080, Mexico;
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional (INP), Mexico City 11340, Mexico
| | - Juan C. Gomez-Verjan
- Dirección de Investigación, Instituto Nacional de Geriatría (INGER), Mexico City 10200, Mexico; (J.C.G.-V.); (N.A.R.-S.)
| | - Nadia A. Rivero-Segura
- Dirección de Investigación, Instituto Nacional de Geriatría (INGER), Mexico City 10200, Mexico; (J.C.G.-V.); (N.A.R.-S.)
| | - Ángel Camarena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City 14080, Mexico;
| | - Ana Karen Torres-Soria
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.V.-O.); (A.K.T.-S.)
| | - Georgina Gonzalez-Avila
- Laboratorio de Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City 14080, Mexico;
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City 14080, Mexico;
| | - Héctor Solís-Chagoyán
- Laboratorio de Neurobiología Cognitiva, Centro de Investigación en Ciencias Cognitivas, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico;
| | - Ruth Jaimez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico; (E.F.-S.); (B.S.R.-M.); (R.J.)
| | | | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City 14080, Mexico;
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
| |
Collapse
|
7
|
Pybus HJ, O'Dea RD, Brook BS. A dynamical model of TGF-β activation in asthmatic airways. MATHEMATICAL MEDICINE AND BIOLOGY : A JOURNAL OF THE IMA 2023; 40:238-265. [PMID: 37285178 DOI: 10.1093/imammb/dqad004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/08/2023]
Abstract
Excessive activation of the regulatory cytokine transforming growth factor $\beta $ (TGF-$\beta $) via contraction of airway smooth muscle (ASM) is associated with the development of asthma. In this study, we develop an ordinary differential equation model that describes the change in density of the key airway wall constituents, ASM and extracellular matrix (ECM), and their interplay with subcellular signalling pathways leading to the activation of TGF-$\beta $. We identify bistable parameter regimes where there are two positive steady states, corresponding to either reduced or elevated TGF-$\beta $ concentration, with the latter leading additionally to increased ASM and ECM density. We associate the former with a healthy homeostatic state and the latter with a diseased (asthmatic) state. We demonstrate that external stimuli, inducing TGF-$\beta $ activation via ASM contraction (mimicking an asthmatic exacerbation), can perturb the system irreversibly from the healthy state to the diseased one. We show that the properties of the stimuli, such as their frequency or strength, and the clearance of surplus active TGF-$\beta $, are important in determining the long-term dynamics and the development of disease. Finally, we demonstrate the utility of this model in investigating temporal responses to bronchial thermoplasty, a therapeutic intervention in which ASM is ablated by applying thermal energy to the airway wall. The model predicts the parameter-dependent threshold damage required to obtain irreversible reduction in ASM content, suggesting that certain asthma phenotypes are more likely to benefit from this intervention.
Collapse
Affiliation(s)
- Hannah J Pybus
- Department of Bioengineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Reuben D O'Dea
- School of Mathematical Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Bindi S Brook
- School of Mathematical Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
8
|
Wang S, Qin S, Cai B, Zhan J, Chen Q. Promising therapeutic mechanism for Chinese herbal medicine in ameliorating renal fibrosis in diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:932649. [PMID: 37522131 PMCID: PMC10376707 DOI: 10.3389/fendo.2023.932649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/22/2023] [Indexed: 08/01/2023] Open
Abstract
Diabetic nephropathy (DN) is one of the most serious chronic microvascular abnormalities of diabetes mellitus and the major cause of uremia. Accumulating evidence has confirmed that fibrosis is a significant pathological feature that contributes to the development of chronic kidney disease in DN. However, the exact mechanism of renal fibrosis in DN is still unclear, which greatly hinders the treatment of DN. Chinese herbal medicine (CHM) has shown efficacy and safety in ameliorating inflammation and albuminuria in diabetic patients. In this review, we outline the underlying mechanisms of renal fibrosis in DN, including oxidative stress (OS) generation and OS-elicited ASK1-p38/JNK activation. Also, we briefly summarize the current status of CHM treating DN by improving renal fibrosis. The treatment of DN by inhibiting ASK1 activation to alleviate renal fibrosis in DN with CHM will promote the discovery of novel therapeutic targets for DN and provide a beneficial therapeutic method for DN.
Collapse
Affiliation(s)
- Shengju Wang
- Department of Nephrology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shuai Qin
- Department of Nephrology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Baochao Cai
- Diabetes Department, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, Zhejiang, China
| | - Jihong Zhan
- Department of Nephrology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Baudo G, Wu S, Massaro M, Liu H, Lee H, Zhang A, Hamilton DJ, Blanco E. Polymer-Functionalized Mitochondrial Transplantation to Fibroblasts Counteracts a Pro-Fibrotic Phenotype. Int J Mol Sci 2023; 24:10913. [PMID: 37446100 PMCID: PMC10342003 DOI: 10.3390/ijms241310913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Fibroblast-to-myofibroblast transition (FMT) leads to excessive extracellular matrix (ECM) deposition-a well-known hallmark of fibrotic disease. Transforming growth factor-β (TGF-β) is the primary cytokine driving FMT, and this phenotypic conversion is associated with mitochondrial dysfunction, notably a metabolic reprogramming towards enhanced glycolysis. The objective of this study was to examine whether the establishment of favorable metabolic phenotypes in TGF-β-stimulated fibroblasts could attenuate FMT. The hypothesis was that mitochondrial replenishment of TGF-β-stimulated fibroblasts would counteract a shift towards glycolytic metabolism, consequently offsetting pro-fibrotic processes. Isolated mitochondria, functionalized with a dextran and triphenylphosphonium (TPP) (Dex-TPP) polymer conjugate, were administered to fibroblasts (MRC-5 cells) stimulated with TGF-β, and effects on bioenergetics and fibrotic programming were subsequently examined. Results demonstrate that TGF-β stimulation of fibroblasts led to FMT, which was associated with enhanced glycolysis. Dex-TPP-coated mitochondria (Dex-TPP/Mt) delivery to TGF-β-stimulated fibroblasts abrogated a metabolic shift towards glycolysis and led to a reduction in reactive oxygen species (ROS) generation. Importantly, TGF-β-stimulated fibroblasts treated with Dex-TPP/Mt had lessened expression of FMT markers and ECM proteins, as well as reduced migration and proliferation. Findings highlight the potential of mitochondrial transfer, as well as other strategies involving functional reinforcement of mitochondria, as viable therapeutic modalities in fibrosis.
Collapse
Affiliation(s)
- Gherardo Baudo
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Suhong Wu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Matteo Massaro
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haoran Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Hyunho Lee
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Dale J. Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Cardiology, Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
10
|
Sun HN, Ren CX, Lee DH, Wang WH, Guo XY, Hao YY, Wang XM, Zhang HN, Xiao WQ, Li N, Cong J, Han YH, Kwon T. PRDX1 negatively regulates bleomycin-induced pulmonary fibrosis via inhibiting the epithelial-mesenchymal transition and lung fibroblast proliferation in vitro and in vivo. Cell Mol Biol Lett 2023; 28:48. [PMID: 37268886 DOI: 10.1186/s11658-023-00460-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/12/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Pulmonary fibrosis is a major category of end-stage changes in lung diseases, characterized by lung epithelial cell damage, proliferation of fibroblasts, and accumulation of extracellular matrix. Peroxiredoxin 1 (PRDX1), a member of the peroxiredoxin protein family, participates in the regulation of the levels of reactive oxygen species in cells and various other physiological activities, as well as the occurrence and development of diseases by functioning as a chaperonin. METHODS Experimental methods including MTT assay, morphological observation of fibrosis, wound healing assay, fluorescence microscopy, flow cytometry, ELISA, western blot, transcriptome sequencing, and histopathological analysis were used in this study. RESULTS PRDX1 knockdown increased ROS levels in lung epithelial cells and promoted epithelial-mesenchymal transition (EMT) through the PI3K/Akt and JNK/Smad signalling pathways. PRDX1 knockout significantly increased TGF-β secretion, ROS production, and cell migration in primary lung fibroblasts. PRDX1 deficiency also increased cell proliferation, cell cycle circulation, and fibrosis progression through the PI3K/Akt and JNK/Smad signalling pathways. BLM treatment induced more severe pulmonary fibrosis in PRDX1-knockout mice, mainly through the PI3K/Akt and JNK/Smad signalling pathways. CONCLUSIONS Our findings strongly suggest that PRDX1 is a key molecule in BLM-induced lung fibrosis progression and acts through modulating EMT and lung fibroblast proliferation; therefore, it may be a therapeutic target for the treatment of BLM-induced lung fibrosis.
Collapse
Affiliation(s)
- Hu-Nan Sun
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China.
| | - Chen-Xi Ren
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Dong Hun Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Wei-Hao Wang
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Xiao-Yu Guo
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Ying-Ying Hao
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Xiao-Ming Wang
- Yabian Academy of Agricultural Science, Longjing, Jilin, 1334000, China
| | - Hui-Na Zhang
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Wan-Qiu Xiao
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Nan Li
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Jie Cong
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Ying-Hao Han
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China.
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 351-33 Neongme-Gil, Ibam-Myeon, Jeongeup-Si, Jeonbuk, 56216, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
11
|
Ligresti G, Raslan AA, Hong J, Caporarello N, Confalonieri M, Huang SK. Mesenchymal cells in the Lung: Evolving concepts and their role in fibrosis. Gene 2023; 859:147142. [PMID: 36603696 PMCID: PMC10068350 DOI: 10.1016/j.gene.2022.147142] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023]
Abstract
Mesenchymal cells in the lung are crucial during development, but also contribute to the pathogenesis of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF), the most common and deadly form of fibrotic interstitial lung diseases. Originally thought to behave as supporting cells for the lung epithelium and endothelium with a singular function of producing basement membrane, mesenchymal cells encompass a variety of cell types, including resident fibroblasts, lipofibroblasts, myofibroblasts, smooth muscle cells, and pericytes, which all occupy different anatomic locations and exhibit diverse homeostatic functions in the lung. During injury, each of these subtypes demonstrate remarkable plasticity and undergo varying capacity to proliferate and differentiate into activated myofibroblasts. Therefore, these cells secrete high levels of extracellular matrix (ECM) proteins and inflammatory cytokines, which contribute to tissue repair, or in pathologic situations, scarring and fibrosis. Whereas epithelial damage is considered the initial trigger that leads to lung injury, lung mesenchymal cells are recognized as the ultimate effector of fibrosis and attempts to better understand the different functions and actions of each mesenchymal cell subtype will lead to a better understanding of why fibrosis develops and how to better target it for future therapy. This review summarizes current findings related to various lung mesenchymal cells as well as signaling pathways, and their contribution to the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Giovanni Ligresti
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US.
| | - Ahmed A Raslan
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Jeongmin Hong
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, US
| | - Marco Confalonieri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, US
| |
Collapse
|
12
|
Yan S, Li M, Liu B, Ma Z, Yang Q. Neutrophil extracellular traps and pulmonary fibrosis: an update. J Inflamm (Lond) 2023; 20:2. [PMID: 36658568 PMCID: PMC9851107 DOI: 10.1186/s12950-023-00329-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Pulmonary fibrosis (PF) is a serious and often fatal illness that occurs in various clinical settings and represents a significant unmet medical need. Increasing evidence indicates that neutrophil extracellular traps (NETs) contribute significantly to the progression of PF. Therefore, understanding the pathways by which NETs contribute to the disease is crucial for developing effective treatments. This review focuses on the formation of NETs and the common mechanisms of NETs in PF.
Collapse
Affiliation(s)
- Suyan Yan
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Meiqi Li
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Baocheng Liu
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Zhenzhen Ma
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Qingrui Yang
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| |
Collapse
|
13
|
Fan G, Liu J, Wu Z, Li C, Zhang Y. Development and validation of the prognostic model based on autophagy-associated genes in idiopathic pulmonary fibrosis. Front Immunol 2022; 13:1049361. [PMID: 36578501 PMCID: PMC9791216 DOI: 10.3389/fimmu.2022.1049361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease. Many studies suggest that autophagy may be related to disease progression and prognosis in IPF. However, the mechanisms involved have not been fully elucidated. Methods We incorporated 232 autophagy-associated genes (AAGs) and two datasets, GSE28042 and GSE27957, from the GEO database. Univariate Cox analysis and least absolute shrinkage and selection operator (LASSO) regression were used to construct the autophagy-associated prognostic model. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to investigate the functions of these autophagy-associated genes. CIBERSORT algorithm was used to calculate the immune cell infiltration between patients in the high-risk score and low-risk score groups. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) was performed to explore the mRNA expression of five genes in the autophagy-associated risk model. Results We constructed a 5-autophagy-associated genes signature based on Univariate Cox analysis and LASSO regression. In our autophagy-associated risk model, IPF patients in the high-risk group demonstrated a poor overall survival rate compared to patients in the low-risk group. For 1-, 2-, and 3-year survival rates, the AUC predictive value of the AAG signature was 0.670, 0.787, and 0.864, respectively. These results were validated in the GSE27957 cohort, confirming the good prognostic effect of our model. GO and KEGG pathway analyses enriched immune-related pathways between the high-risk and low-risk groups. And there was also a significant difference in immune cell infiltration between two groups. And the results of qRT-PCR showed that the expression levels of FOXO1, IRGM, MYC, and PRKCQ were significantly decreased in the Peripheral Blood Mononuclear Cell (PBMC) of IPF patient samples. Conclusion Our study constructed and validated an autophagy-associated risk model based on MYC, MAPK1, IRGM, PRKCQ, and FOXO1. And those five genes may influence the progression of IPF by regulating immune responses and immune cells.
Collapse
Affiliation(s)
- Guoqing Fan
- Department of Respiratory Medicine and Critical Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China,Graduate School of Peking Union Medical College, Beijing, China
| | - Jingjing Liu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhen Wu
- Department of Respiratory & Critical Care Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Caiyu Li
- Department of Respiratory & Critical Care Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ying Zhang
- Department of Respiratory & Critical Care Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China,*Correspondence: Ying Zhang,
| |
Collapse
|
14
|
Lin S, Lin R, Zhang H, Xu Q, He Y. Peripheral vascular remodeling during ischemia. Front Pharmacol 2022; 13:1078047. [DOI: 10.3389/fphar.2022.1078047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
About 230 million people worldwide suffer from peripheral arterial disease (PAD), and the prevalence is increasing year by year. Multiple risk factors, including smoking, dyslipidemia, diabetes, and hypertension, can contribute to the development of PAD. PAD is typically characterized by intermittent claudication and resting pain, and there is a risk of severe limb ischemia, leading to major adverse limb events, such as amputation. Currently, a major progress in the research field of the pathogenesis of vascular remodeling, including atherosclerosis and neointima hyperplasia has been made. For example, the molecular mechanisms of endothelial dysfunction and smooth muscle phenotype switching have been described. Interestingly, a series of focused studies on fibroblasts of the vessel wall has demonstrated their impact on smooth muscle proliferation and even endothelial function via cell-cell communications. In this review, we aim to focus on the functional changes of peripheral arterial cells and the mechanisms of the pathogenesis of PAD. At the same time, we summarize the progress of the current clinical treatment and potential therapeutic methods for PAD and shine a light on future perspectives.
Collapse
|
15
|
Karadeniz H, Avanoğlu Güler A, Özger HS, Yıldız PA, Erbaş G, Bozdayı G, Deveci Bulut T, Gülbahar Ö, Yapar D, Küçük H, Öztürk MA, Tufan A. The Prognostic Value of Lung Injury and Fibrosis Markers, KL-6,
TGF-β1, FGF-2 in COVID-19 Patients. Biomark Insights 2022; 17:11772719221135443. [PMCID: PMC9643117 DOI: 10.1177/11772719221135443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/11/2022] [Indexed: 11/11/2022] Open
Abstract
Background: Biomarkers of lung injury and interstitial fibrosis give insight about the
extent of involvement and prognosis in well-known interstitial lung diseases
(ILD). Serum Krebs von den Lungen-6 (KL-6) reflects direct alveolar injury
and, transforming growth factor-beta1 (TGF-β1) and fibroblast growth
factor-2 (FGF-2) are principal mediators of fibrosis in ILD and in almost
all fibrotic diseases. In this sense, we aimed to assess associations of
these biomarkers with traditional inflammatory markers and clinical course
of COVID-19. Methods: Patients with COVID-19 who had confirmed diagnosis with SARS-CoV-2 nucleic
acid RT-PCR were enrolled and followed up prospectively with a standardized
approach one month after diagnosis. Patients were divided into severe and
non-severe groups according to National Institutes of Health criteria.
Outcome was assessed for the requirement of intensive care unit (ICU)
admission, long term respiratory support and death. Blood samples were
collected at enrollment and serum levels of KL-6, TGF-β1, FGF-2 were
determined by ELISA. Association between these markers with other prognostic
markers and prognosis were analyzed. Results: Overall 31 severe and 28 non-severe COVID-19 patients were enrolled and were
compared with healthy control subjects (n = 30). Serum KL-6 levels in
COVID-19 patients were significantly higher (median [IQR]; 11.54 [4.86] vs
8.54 [3.98] ng/mL, P = .001] and FGF-2 levels were lower
(median [IQR]; 76.84 [98.2] vs 101.62 [210.6] pg/mL) compared to healthy
control group. A significant correlation was found between KL-6 values and
CRP, fibrinogen, d-dimer and lymphocyte counts. However, we did not
find an association between these markers and subsequent severity of
COVID-19, mortality and long-term prognosis. Conclusions: Serum KL-6 levels were significantly elevated at the diagnosis of COVID-19
and correlated well with the other traditional prognostic inflammatory
markers. Serum levels of principal fibrosis mediators, TGF-β1, FGF-2, were
not elevated at diagnosis of COVID-19, therefore did not help to anticipate
long term prognosis.
Collapse
Affiliation(s)
- Hazan Karadeniz
- Division of Rheumatology, Department of
Internal Medicine, Gazi University Faculty of Medicine, Ankara, Turkey,Hazan Karadeniz, Department of Internal
Medicine, Division of Rheumatology, Gazi University Faculty of Medicine,
Bahriucok Street, Ankara 06100, Turkey.
| | - Aslıhan Avanoğlu Güler
- Division of Rheumatology, Department of
Internal Medicine, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Hasan Selçuk Özger
- Department of Infectious Disease, Gazi
University Faculty of Medicine, Ankara, Turkey
| | - Pınar Aysert Yıldız
- Department of Infectious Disease, Gazi
University Faculty of Medicine, Ankara, Turkey
| | - Gonca Erbaş
- Department of Radiology, Gazi
University Faculty of Medicine, Ankara, Turkey
| | - Gülendam Bozdayı
- Department of Medical Microbiology,
Gazi University Faculty of Medicine, Ankara, Turkey
| | - Tuba Deveci Bulut
- Department of Biochemistry, Gazi
University Faculty of Medicine, Ankara, Turkey
| | - Özlem Gülbahar
- Department of Biochemistry, Gazi
University Faculty of Medicine, Ankara, Turkey
| | - Dilek Yapar
- Department of Public Health and
Biostatistics Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Hamit Küçük
- Division of Rheumatology, Department of
Internal Medicine, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Mehmet Akif Öztürk
- Division of Rheumatology, Department of
Internal Medicine, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Abdurrahman Tufan
- Division of Rheumatology, Department of
Internal Medicine, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
16
|
Sun C, Tian X, Jia Y, Yang M, Li Y, Fernig DG. Functions of exogenous FGF signals in regulation of fibroblast to myofibroblast differentiation and extracellular matrix protein expression. Open Biol 2022; 12:210356. [PMID: 36102060 PMCID: PMC9471990 DOI: 10.1098/rsob.210356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Fibroblasts are widely distributed cells found in most tissues and upon tissue injury, they are able to differentiate into myofibroblasts, which express abundant extracellular matrix (ECM) proteins. Overexpression and unordered organization of ECM proteins cause tissue fibrosis in damaged tissue. Fibroblast growth factor (FGF) family proteins are well known to promote angiogenesis and tissue repair, but their activities in fibroblast differentiation and fibrosis have not been systematically reviewed. Here we summarize the effects of FGFs in fibroblast to myofibroblast differentiation and ECM protein expression and discuss the underlying potential regulatory mechanisms, to provide a basis for the clinical application of recombinant FGF protein drugs in treatment of tissue damage.
Collapse
Affiliation(s)
- Changye Sun
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Xiangqin Tian
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Yangyang Jia
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Mingming Yang
- Department of Cardiology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, People's Republic of China
| | - Yong Li
- Department of Biochemistry, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - David G Fernig
- Department of Biochemistry, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| |
Collapse
|
17
|
Golgin Subfamily A Member 5 Is Essential for Production of Extracellular Matrix Proteins during TGF-β1-Induced Periodontal Ligament-Fibroblastic Differentiation. Stem Cells Int 2022; 2022:3273779. [PMID: 35879965 PMCID: PMC9308542 DOI: 10.1155/2022/3273779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/05/2022] [Indexed: 11/21/2022] Open
Abstract
Human periodontal ligament stem cells (hPDLSCs) can be differentiated into periodontal ligament- (PDL-) fibroblastic progenitors by treatment with low concentrations of transforming growth factor beta 1 (TGF-β1). Although much is known about the profibrotic effects of TGF-β1, the molecular mechanisms mediating the activation of fibroblasts in periodontal ligament-fibroblastic differentiation are not well known. Our study was to investigate the mechanism of the fibroblastic process in the periodontal ligament differentiation of hPDLSCs through the discovery of novel markers. One of the monoclonal antibodies previously established through decoy immunization was the anti-LG11 antibody, which recognized Golgi subfamily A member 5 (GOLGA5) as a PDL-fibroblastic progenitor-specific antigen. GOLGA5/LG11 was significantly upregulated in TGF-β1-induced PDL-fibroblastic progenitors and accumulated in the PDL region of the tooth root. GOLGA5 plays a role in vesicle tethering and docking between the endoplasmic reticulum and the Golgi apparatus. siRNA-mediated depletion of endogenous GOLGA5 upregulated in TGF-β1-induced PDL-fibroblastic progenitors resulted in downregulation of representative PDL-fibroblastic markers and upregulation of osteoblast markers. When the TGF-β1 signaling pathway was blocked or GOLGA5 was depleted by siRNA, the levels of extracellular matrix (ECM) proteins, such as type I collagen and fibronectin, decreased in PDL-fibroblastic progenitors. In addition, Golgi structures in the perinuclear region underwent fragmentation under these conditions. These results suggest that GOLGA5/LG11 is a PDL-fibroblastic marker with functional importance in ECM protein production and secretion, which are important processes in PDL-fibroblastic differentiation.
Collapse
|
18
|
Ma H, Liu S, Li S, Xia Y. Targeting Growth Factor and Cytokine Pathways to Treat Idiopathic Pulmonary Fibrosis. Front Pharmacol 2022; 13:918771. [PMID: 35721111 PMCID: PMC9204157 DOI: 10.3389/fphar.2022.918771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/06/2022] [Indexed: 02/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease of unknown origin that usually results in death from secondary respiratory failure within 2–5 years of diagnosis. Recent studies have identified key roles of cytokine and growth factor pathways in the pathogenesis of IPF. Although there have been numerous clinical trials of drugs investigating their efficacy in the treatment of IPF, only Pirfenidone and Nintedanib have been approved by the FDA. However, they have some major limitations, such as insufficient efficacy, undesired side effects and poor pharmacokinetic properties. To give more insights into the discovery of potential targets for the treatment of IPF, this review provides an overview of cytokines, growth factors and their signaling pathways in IPF, which have important implications for fully exploiting the therapeutic potential of targeting cytokine and growth factor pathways. Advances in the field of cytokine and growth factor pathways will help slow disease progression, prolong life, and improve the quality of life for IPF patients in the future.
Collapse
Affiliation(s)
- Hongbo Ma
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shengming Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shanrui Li
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yong Xia
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
| |
Collapse
|
19
|
Mesenchymal stem cells exert their anti-asthmatic effects through macrophage modulation in a murine chronic asthma model. Sci Rep 2022; 12:9811. [PMID: 35697721 PMCID: PMC9192777 DOI: 10.1038/s41598-022-14027-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 05/31/2022] [Indexed: 12/11/2022] Open
Abstract
Despite numerous previous studies, the full action mechanism of the pathogenesis of asthma remains undiscovered, and the need for further investigation is increasing in order to identify more effective target molecules. Recent attempts to develop more efficacious treatments for asthma have incorporated mesenchymal stem cell (MSC)-based cell therapies. This study aimed to evaluate the anti-asthmatic effects of MSCs primed with Liproxstatin-1, a potent ferroptosis inhibitor. In addition, we sought to examine the changes within macrophage populations and their characteristics in asthmatic conditions. Seven-week-old transgenic mice, constitutively overexpressing lung-specific interleukin (IL)-13, were used to simulate chronic asthma. Human umbilical cord-derived MSCs (hUC-MSCs) primed with Liproxstatin-1 were intratracheally administered four days prior to sampling. IL-13 transgenic mice demonstrated phenotypes of chronic asthma, including severe inflammation, goblet cell hyperplasia, and subepithelial fibrosis. Ly6C+M2 macrophages, found within the pro-inflammatory CD11c+CD11b+ macrophages, were upregulated and showed a strong correlation with lung eosinophil counts. Liproxstatin-1-primed hUC-MSCs showed enhanced ability to downregulate the activation of T helper type 2 cells compared to naïve MSCs in vitro and reduced airway inflammation, particularly Ly6C+M2 macrophages population, and fibrosis in vivo. In conclusion, intratracheal administration is an effective method of MSC delivery, and macrophages hold great potential as an additional therapeutic target for asthma.
Collapse
|
20
|
Liu G, Li S, Zhang N, Wei N, Wang M, Liu J, Xu Y, Li Y, Sun Q, Li Y, Li F, Yu P, Liu M, Wang Y, Zhai H, Wang Y. Sequential grade evaluation method exploration of Exocarpium Citri Grandis (Huajuhong) decoction pieces based on "network prediction → grading quantization → efficacy validation". JOURNAL OF ETHNOPHARMACOLOGY 2022; 291:115149. [PMID: 35231589 DOI: 10.1016/j.jep.2022.115149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/08/2022] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Exocarpium Citri Grandis (Huajuhong) is an authentic Chinese materia medica with excellent curative effects on relieving cough and reducing phlegm, which has been reputed as "Southern Ginseng" in China for a long history. AIM OF THE STUDY To establish a sequential grade evaluation method with strong operability and controllable quality for Huajuhong decoction pieces. MATERIALS AND METHODS (1) Indicators of ingredients and bio-effects were predicted by network pharmacology, and the potential pharmacodynamic ingredients and key targets were analyzed integrating screening results and literatures. (2) 45 batches of Huajuhong decoction pieces from different producing areas were collected and graded by original plant, planting place, and harvesting time. The chemical indicators determination of Huajuhong decoction pieces was conducted by Ultra Performance Liquid Chromatography (UPLC). (3) 112 rats with idiopathic pulmonary fibrosis (IPF) model were used to evaluated the efficacy within graded groups. RESULTS (1) There are 22 key targets corresponding to 20 potential ingredients related to immunity and inflammation pathways for Huajuhong. Naringin and rhoifolin were chosen as the chemical indicators, and IL-6, IL-8, MCP-1, MIP-1α, TNF-α, TGF-β1 were selected as bio-indicators for different grades of Huajuhong decoction pieces. (2) The contents of the naringin and rhoifolin can reflect the quality of different grades of Huajuhong decoction pieces. (3) The efficacy of different grades of Huajuhong decoction pieces can delay the progression of IPF in varying degrees via the selected bio-indicators' pathways. CONCLUSIONS This sequential grading evaluation method is an attempt to apply systems pharmacology which integrates network pharmacology, quantitative chemical and experiments on animals to the classification of TCM decoction pieces. Combining the concepts of traditional theory and modern technology to explain the complex grading mechanism of TCM decoction pieces is worth popularizing and applying.
Collapse
Affiliation(s)
- Guoxiu Liu
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Siyu Li
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Nan Zhang
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Namin Wei
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mengxin Wang
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jiao Liu
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yan Xu
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yanan Li
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Qianqian Sun
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yixuan Li
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Feng Li
- Huazhou Huajuhong Medicinal Materials Development Co. LTD, Guangdong, 525199, China
| | - Pinhao Yu
- Huazhou Huajuhong Medicinal Materials Development Co. LTD, Guangdong, 525199, China
| | - Mengyu Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanping Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Huaqiang Zhai
- Standardization Research Center of Traditional Chinese Medicine Dispensing, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Yongyan Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| |
Collapse
|
21
|
Kou L, Kou P, Luo G, Wei S. Progress of Statin Therapy in the Treatment of Idiopathic Pulmonary Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6197219. [PMID: 35345828 PMCID: PMC8957418 DOI: 10.1155/2022/6197219] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/24/2022] [Indexed: 11/18/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a type of interstitial lung disease (ILD) characterized by the proliferation of fibroblasts and aberrant accumulation of extracellular matrix. These changes are accompanied by structural destruction of the lung tissue and the progressive decline of pulmonary function. In the past few decades, researchers have investigated the pathogenesis of IPF and sought a therapeutic approach for its treatment. Some studies have shown that the occurrence of IPF is related to pulmonary inflammatory injury; however, its specific etiology and pathogenesis remain unknown, and no effective treatment, with the exception of lung transplantation, has been identified yet. Several basic science and clinical studies in recent years have shown that statins, the traditional lipid-lowering drugs, exert significant antifibrotic effects, which can delay the progression of IPF and impairment of pulmonary function. This article is aimed at summarizing the current understanding of the pathogenesis of IPF, the progress of research on the use of statins in IPF models and clinical trials, and its main molecular targets.
Collapse
Affiliation(s)
- Leiya Kou
- Department of Respiratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, China
- Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Pei Kou
- Department of Medical Record, Wuhan No. 1 Hospital, Wuhan 430022, China
| | - Guangwei Luo
- Department of Respiratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
22
|
Robinson S, Parigoris E, Chang J, Hecker L, Takayama S. Contracting scars from fibrin drops. Integr Biol (Camb) 2022; 14:1-12. [PMID: 35184163 PMCID: PMC8934703 DOI: 10.1093/intbio/zyac001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Indexed: 11/13/2022]
Abstract
This paper describes a microscale fibroplasia and contraction model that is based on fibrin-embedded lung fibroblasts and provides a convenient visual readout of fibrosis. Cell-laden fibrin microgel drops are formed by aqueous two-phase microprinting. The cells deposit extracellular matrix (ECM) molecules such as collagen while fibrin is gradually degraded. Ultimately, the cells contract the collagen-rich matrix to form a compact cell-ECM spheroid. The size of the spheroid provides the visual readout of the extent of fibroplasia. Stimulation of this wound-healing model with the profibrotic cytokine TGF-β1 leads to an excessive scar formation response that manifests as increased collagen production and larger cell-ECM spheroids. Addition of drugs also shifted the scarring profile: the FDA-approved fibrosis drugs (nintedanib and pirfenidone) and a PAI-1 inhibitor (TM5275) significantly reduced cell-ECM spheroid size. Not only is the assay useful for evaluation of antifibrotic drug effects, it is relatively sensitive; one of the few in vitro fibroplasia assays that can detect pirfenidone effects at submillimolar concentrations. Although this paper focuses on lung fibrosis, the approach opens opportunities for studying a broad range of fibrotic diseases and for evaluating antifibrotic therapeutics.
Collapse
Affiliation(s)
| | - Eric Parigoris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA,The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jonathan Chang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA,The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Louise Hecker
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA,The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
23
|
Saleh M, Fotook Kiaei SZ, Kavianpour M. Application of Wharton jelly-derived mesenchymal stem cells in patients with pulmonary fibrosis. Stem Cell Res Ther 2022; 13:71. [PMID: 35168663 PMCID: PMC8845364 DOI: 10.1186/s13287-022-02746-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Pulmonary fibrosis is a devastating disease that eventually leads to death and respiratory failure. Despite the wide range of drugs, including corticosteroids, endothelin antagonist, and pirfenidone, there is no effective treatment, and the only main goal of treatment is to alleviate the symptoms as much as possible to slow down the progression of the disease and improve the quality of life. Lung transplantation may be a treatment option for a few people if pulmonary fibrosis develops and there is no established treatment. Pulmonary fibrosis caused by the COVID19 virus is another problem that we face in most patients despite the efforts of the international medical communities. Therefore, achieving alternative treatment for patients is a great success. Today, basic research using stem cells on pulmonary fibrosis has published promising results. New stem cell-based therapies can be helpful in patients with pulmonary fibrosis. Wharton jelly-derived mesenchymal stem cells are easily isolated in large quantities and made available for clinical trials without causing ethical problems. These cells have higher flexibility and proliferation potential than other cells isolated from different sources and differentiated into various cells in laboratory environments. More clinical trials are needed to determine the safety and efficacy of these cells. This study will investigate the cellular and molecular mechanisms and possible effects of Wharton jelly-derived mesenchymal stem cells in pulmonary fibrosis.
Collapse
Affiliation(s)
- Mahshid Saleh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyedeh Zahra Fotook Kiaei
- Department of Pulmonary and Critical Care, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maria Kavianpour
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Xia Y, Cheng M, Hu Y, Li M, Shen L, Ji X, Cui X, Liu X, Wang W, Gao H. Combined transcriptomic and lipidomic analysis of D-4F ameliorating bleomycin-induced pulmonary fibrosis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1424. [PMID: 34733976 PMCID: PMC8506780 DOI: 10.21037/atm-21-3777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/26/2021] [Indexed: 11/06/2022]
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease that leads to respiratory failure, and for which there is no effective treatment. Apolipoprotein A-1 (ApoA-1) has been reported to ameliorate the bleomycin (BLM)-induced IPF model. Methods To examine the function of D-4F, an ApoA-1 mimetic polypeptide, in IPF, we used an in-vivo BLM-induced model. We assigned mice into the following 3 groups: the Blank Group (BLK Group), the Bleomycin Treatment Group (Model Group), and the D-4F Interference Group (Inter Group). The BLM-induced fibrosis was examined by hematoxylin and eosin, Masson’s trichrome (M-T) staining and immunohistochemical staining. An untargeted lipidomic and transcriptomic analysis were used to examine the function of D-4F. Results There were 35 differentially altered lipids (DALs) in the BLK, Model and Inter Groups. A Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that glycerophospholipid metabolism was the most highly enriched of the 35 DALs. There were 99 differentially expressed genes (DEGs) in the BLK, Model and Inter Groups. The enriched KEGG pathway analysis showed that the mitogen-activated protein kinase (MAPK) pathway was 1 of the top 10 pathways. The results of the untargeted lipidomic and transcriptomic analysis showed that phospholipase A2 group 4c (Pla2g4c) was a crucial gene in both the MAPK pathway and glycerophospholipid metabolism. Pla2g4c was increased in the Model Group but decreased in the Inter Group. Conclusions It may be that D-4F prevented the BLM-induced pulmonary fibrosis model by inhibiting the expression of pla2g4c. Our findings suggest that D-4F may be a potential treatment of IPF.
Collapse
Affiliation(s)
- Yong Xia
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Shandong provincial Key Laboratory of Cardiovascular Proteomics, Shandong University, Jinan, China
| | - Mei Cheng
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Shandong provincial Key Laboratory of Cardiovascular Proteomics, Shandong University, Jinan, China
| | - Yanyan Hu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Shandong provincial Key Laboratory of Cardiovascular Proteomics, Shandong University, Jinan, China
| | - Man Li
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Shandong provincial Key Laboratory of Cardiovascular Proteomics, Shandong University, Jinan, China
| | - Lin Shen
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Shandong provincial Key Laboratory of Cardiovascular Proteomics, Shandong University, Jinan, China
| | - Xiang Ji
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Xiaopei Cui
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Shandong provincial Key Laboratory of Cardiovascular Proteomics, Shandong University, Jinan, China
| | - Xiangju Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Shandong provincial Key Laboratory of Cardiovascular Proteomics, Shandong University, Jinan, China
| | - Weiling Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Shandong provincial Key Laboratory of Cardiovascular Proteomics, Shandong University, Jinan, China
| | - Haiqing Gao
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Shandong provincial Key Laboratory of Cardiovascular Proteomics, Shandong University, Jinan, China
| |
Collapse
|
25
|
Cisterna B, Costanzo M, Lacavalla MA, Galiè M, Angelini O, Tabaracci G, Malatesta M. Low Ozone Concentrations Differentially Affect the Structural and Functional Features of Non-Activated and Activated Fibroblasts In Vitro. Int J Mol Sci 2021; 22:10133. [PMID: 34576295 PMCID: PMC8466365 DOI: 10.3390/ijms221810133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/13/2022] Open
Abstract
Oxygen-ozone (O2-O3) therapy is increasingly applied as a complementary/adjuvant treatment for several diseases; however, the biological mechanisms accounting for the efficacy of low O3 concentrations need further investigations to understand the possibly multiple effects on the different cell types. In this work, we focused our attention on fibroblasts as ubiquitous connective cells playing roles in the body architecture, in the homeostasis of tissue-resident cells, and in many physiological and pathological processes. Using an established human fibroblast cell line as an in vitro model, we adopted a multimodal approach to explore a panel of cell structural and functional features, combining light and electron microscopy, Western blot analysis, real-time quantitative polymerase chain reaction, and multiplex assays for cytokines. The administration of O2-O3 gas mixtures induced multiple effects on fibroblasts, depending on their activation state: in non-activated fibroblasts, O3 stimulated proliferation, formation of cell surface protrusions, antioxidant response, and IL-6 and TGF-β1 secretion, while in LPS-activated fibroblasts, O3 stimulated only antioxidant response and cytokines secretion. Therefore, the low O3 concentrations used in this study induced activation-like responses in non-activated fibroblasts, whereas in already activated fibroblasts, the cell protective capability was potentiated.
Collapse
Affiliation(s)
- Barbara Cisterna
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy; (B.C.); (M.C.); (M.A.L.); (M.G.)
| | - Manuela Costanzo
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy; (B.C.); (M.C.); (M.A.L.); (M.G.)
| | - Maria Assunta Lacavalla
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy; (B.C.); (M.C.); (M.A.L.); (M.G.)
| | - Mirco Galiè
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy; (B.C.); (M.C.); (M.A.L.); (M.G.)
| | - Osvaldo Angelini
- San Rocco Clinic, Via Monsignor G.V. Moreni 95, I-25018 Montichari, Italy; (O.A.); (G.T.)
| | - Gabriele Tabaracci
- San Rocco Clinic, Via Monsignor G.V. Moreni 95, I-25018 Montichari, Italy; (O.A.); (G.T.)
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy; (B.C.); (M.C.); (M.A.L.); (M.G.)
| |
Collapse
|
26
|
Ye Z, Hu Y. TGF‑β1: Gentlemanly orchestrator in idiopathic pulmonary fibrosis (Review). Int J Mol Med 2021; 48:132. [PMID: 34013369 PMCID: PMC8136122 DOI: 10.3892/ijmm.2021.4965] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/29/2021] [Indexed: 01/09/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a worldwide disease characterized by the chronic and irreversible decline of lung function. Currently, there is no drug to successfully treat the disease except for lung transplantation. Numerous studies have been devoted to the study of the fibrotic process of IPF and findings showed that transforming growth factor‑β1 (TGF‑β1) plays a central role in the development of IPF. TGF‑β1 promotes the fibrotic process of IPF through various signaling pathways, including the Smad, MAPK, and ERK signaling pathways. There are intersections between these signaling pathways, which provide new targets for researchers to study new drugs. In addition, TGF‑β1 can affect the fibrosis process of IPF by affecting oxidative stress, epigenetics and other aspects. Most of the processes involved in TGF‑β1 promote IPF, but TGF‑β1 can also inhibit it. This review discusses the role of TGF‑β1 in IPF.
Collapse
Affiliation(s)
- Zhimin Ye
- Department of Pathology, Basic Medical School, Central South University, Changsha, Hunan 410006, P.R. China
| | - Yongbin Hu
- Department of Pathology, Basic Medical School, Central South University, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
27
|
John AE, Joseph C, Jenkins G, Tatler AL. COVID-19 and pulmonary fibrosis: A potential role for lung epithelial cells and fibroblasts. Immunol Rev 2021; 302:228-240. [PMID: 34028807 PMCID: PMC8237078 DOI: 10.1111/imr.12977] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic rapidly spread around the world following the first reports in Wuhan City, China in late 2019. The disease, caused by the novel SARS-CoV-2 virus, is primarily a respiratory condition that can affect numerous other bodily systems including the cardiovascular and gastrointestinal systems. The disease ranges in severity from asymptomatic through to severe acute respiratory distress requiring intensive care treatment and mechanical ventilation, which can lead to respiratory failure and death. It has rapidly become evident that COVID-19 patients can develop features of interstitial pulmonary fibrosis, which in many cases persist for as long as we have thus far been able to follow the patients. Many questions remain about how such fibrotic changes occur within the lung of COVID-19 patients, whether the changes will persist long term or are capable of resolving, and whether post-COVID-19 pulmonary fibrosis has the potential to become progressive, as in other fibrotic lung diseases. This review brings together our existing knowledge on both COVID-19 and pulmonary fibrosis, with a particular focus on lung epithelial cells and fibroblasts, in order to discuss common pathways and processes that may be implicated as we try to answer these important questions in the months and years to come.
Collapse
Affiliation(s)
- Alison E. John
- Nottingham NIHR Respiratory Biomedical Research CentreUniversity of NottinghamNottinghamUK
- National Heart and Lung InstituteImperial CollegeLondonUK
| | - Chitra Joseph
- Nottingham NIHR Respiratory Biomedical Research CentreUniversity of NottinghamNottinghamUK
| | - Gisli Jenkins
- Nottingham NIHR Respiratory Biomedical Research CentreUniversity of NottinghamNottinghamUK
- National Heart and Lung InstituteImperial CollegeLondonUK
| | - Amanda L. Tatler
- Nottingham NIHR Respiratory Biomedical Research CentreUniversity of NottinghamNottinghamUK
| |
Collapse
|
28
|
Zhang Y, Liu Z, Wang K, Lu S, Fan S, Xu L, Cai B. Macrophage migration inhibitory factor regulates joint capsule fibrosis by promoting TGF-β1 production in fibroblasts. Int J Biol Sci 2021; 17:1837-1850. [PMID: 33994866 PMCID: PMC8120472 DOI: 10.7150/ijbs.57025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/09/2021] [Indexed: 11/05/2022] Open
Abstract
Joint capsule fibrosis caused by excessive inflammation results in post-traumatic joint contracture (PTJC). Transforming growth factor (TGF)-β1 plays a key role in PTJC by regulating fibroblast functions, however, cytokine-induced TGF-β1 expression in specific cell types remains poorly characterized. Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in inflammation- and fibrosis-associated pathophysiology. In this study, we investigated whether MIF can facilitate TGF-β1 production from fibroblasts and regulate joint capsule fibrosis following PTJC. Our data demonstrated that MIF and TGF-β1 significantly increased in fibroblasts of injured rat posterior joint capsules. Treatment the lesion sites with MIF inhibitor 4-Iodo-6-phenylpyrimidine (4-IPP) reduced TGF-β1 production and relieved joint capsule inflammation and fibrosis. In vitro, MIF facilitated TGF-β1 expression in primary joint capsule fibroblasts by activating mitogen-activated protein kinase (MAPK) (P38, ERK) signaling through coupling with membrane surface receptor CD74, which in turn affected fibroblast functions and promoted MIF production. Our results reveal a novel function of trauma-induced MIF in the occurrence and development of joint capsule fibrosis. Further investigation of the underlying mechanism may provide potential therapeutic targets for PTJC.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,Shanghai Key Laboratory of Orthopedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai 200011, China
| | - Kexin Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Shenji Lu
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shuai Fan
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Lili Xu
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Bin Cai
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
29
|
Al-Habeeb F, Aloufi N, Traboulsi H, Liu X, Nair P, Haston C, Azuelos I, Huang SK, White ES, Gallouzi IE, Di Marco S, Eidelman DH, Baglole CJ. Human antigen R promotes lung fibroblast differentiation to myofibroblasts and increases extracellular matrix production. J Cell Physiol 2021; 236:6836-6851. [PMID: 33855709 DOI: 10.1002/jcp.30380] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 01/12/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disease of progressive scarring caused by excessive extracellular matrix (ECM) deposition and activation of α-SMA-expressing myofibroblasts. Human antigen R (HuR) is an RNA binding protein that promotes protein translation. Upon translocation from the nucleus to the cytoplasm, HuR functions to stabilize messenger RNA (mRNA) to increase protein levels. However, the role of HuR in promoting ECM production, myofibroblast differentiation, and lung fibrosis is unknown. Human lung fibroblasts (HLFs) treated with transforming growth factor β1 (TGF-β1) showed a significant increase in translocation of HuR from the nucleus to the cytoplasm. TGF-β-treated HLFs that were transfected with HuR small interfering RNA had a significant reduction in α-SMA protein as well as the ECM proteins COL1A1, COL3A, and FN1. HuR was also bound to mRNA for ACTA2, COL1A1, COL3A1, and FN. HuR knockdown affected the mRNA stability of ACTA2 but not that of the ECM genes COL1A1, COL3A1, or FN. In mouse models of pulmonary fibrosis, there was higher cytoplasmic HuR in lung structural cells compared to control mice. In human IPF lungs, there was also more cytoplasmic HuR. This study is the first to show that HuR in lung fibroblasts controls their differentiation to myofibroblasts and consequent ECM production. Further research on HuR could assist in establishing the basis for the development of new target therapy for fibrotic diseases, such as IPF.
Collapse
Affiliation(s)
- Fatmah Al-Habeeb
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Noof Aloufi
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Departments of Pathology, McGill University, Montreal, Quebec, Canada
| | - Hussein Traboulsi
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Xingxing Liu
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Parameswaran Nair
- Department of Medicine, McMaster University & St Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Christina Haston
- Department of Computer Science, Mathematics, Physics and Statistics, University of British Columbia, British Columbia, Canada
| | - Ilan Azuelos
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Steven K Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Imed E Gallouzi
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Faculty of Medicine, Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - Sergio Di Marco
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Faculty of Medicine, Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - David H Eidelman
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Carolyn J Baglole
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Departments of Pathology, McGill University, Montreal, Quebec, Canada.,Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
30
|
Damanik FFR, Verkoelen N, van Blitterswijk C, Rotmans J, Moroni L. Control Delivery of Multiple Growth Factors to Actively Steer Differentiation and Extracellular Matrix Protein Production. Adv Biol (Weinh) 2021; 5:e2000205. [PMID: 33751850 DOI: 10.1002/adbi.202000205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/16/2021] [Indexed: 12/21/2022]
Abstract
In tissue engineering, biomaterials have been used to steer the host response. This determines the outcome of tissue regeneration, which is modulated by multiple growth factors (GFs). Hence, a sustainable delivery system for GFs is necessary to control tissue regeneration actively. A delivery technique of single and multiple GF combinations, using a layer-by-layer (LBL) procedure to improve tissue remodeling, is developed. TGF-β1, PDGF-ββ, and IGF-1 are incorporated on tailor-made polymeric rods, which could be used as a tool for potential tissue engineering applications, such as templates to induce the formation of in situ tissue engineered blood vessels (TEBVs). Cell response is analyzed in vitro using rat and human dermal fibroblasts for cellular proliferation, fibroblast differentiation, and extracellular matrix (ECM) protein synthesis. Results revealed a higher loading efficiency and control release of GFs incorporated on chloroform and oxygen plasma-activated (COX) rods. Single PDGF-ββ and IGF-1 release, and dual release with TGF-β1 from COX rods, showed higher cell proliferation when compared to COX rods alone. A substantial increase in α-smooth muscle actin (α-SMA) is also observed in GF releasing COX rods, with TGF-β1 COX rods providing the most pronounced differentiation. A significant increase in collagen and elastin synthesis is observed on all GF releasing COX rods compared to control, with COX rods releasing TGF-β1 and IGF-1 providing the highest secretion. TGF-β1 and IGF-1 releasing COX rods induced higher Glycosaminoglycan (GAG)/DNA amounts than the other GF releasing COX rods. As PDGF-ββ and TGF-β1/PDGF-ββ COX rods displayed the highest fibroblast attachment, these rods provided the highest total collagen and elastin production. The attractive results from efficiently incorporating single and multiple GFs on COX rods and their sustainable release to steer cellular behavior suggest a promising route to enrich the formation of in situ engineered tissues.
Collapse
Affiliation(s)
- Febriyani F R Damanik
- Tissue Regeneration Department, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Drienerlolaan 5, Zuidhorst 145, Enschede, NB, 7522, The Netherlands.,Complex Tissue Regeneration Department, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitsingel 40, Maastricht, 6229 ER, The Netherlands
| | - Niels Verkoelen
- Complex Tissue Regeneration Department, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitsingel 40, Maastricht, 6229 ER, The Netherlands
| | - Clemens van Blitterswijk
- Tissue Regeneration Department, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Drienerlolaan 5, Zuidhorst 145, Enschede, NB, 7522, The Netherlands.,Complex Tissue Regeneration Department, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitsingel 40, Maastricht, 6229 ER, The Netherlands
| | - Joris Rotmans
- Department of Nephrology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Lorenzo Moroni
- Tissue Regeneration Department, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Drienerlolaan 5, Zuidhorst 145, Enschede, NB, 7522, The Netherlands.,Complex Tissue Regeneration Department, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitsingel 40, Maastricht, 6229 ER, The Netherlands
| |
Collapse
|
31
|
Berhan A, Harris T, Jaffar J, Jativa F, Langenbach S, Lönnstedt I, Alhamdoosh M, Ng M, Lee P, Westall G, Wilson N, Wilson M, Stewart AG. Cellular Microenvironment Stiffness Regulates Eicosanoid Production and Signaling Pathways. Am J Respir Cell Mol Biol 2021; 63:819-830. [PMID: 32926636 DOI: 10.1165/rcmb.2020-0227oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pathological changes in the biomechanical environment are implicated in the progression of idiopathic pulmonary fibrosis (IPF). Stiffened matrix augments fibroblast proliferation and differentiation and activates TGF-β1 (transforming growth factor-β1). Stiffened matrix impairs the synthesis of the antifibrogenic lipid mediator prostaglandin E2 (PGE2) and reduces the expression of the rate-limiting prostanoid biosynthetic enzyme cyclooxygenase-2 (COX-2). We now show that prostaglandin E synthase (PTGES), the final enzyme in the PGE2 biosynthetic pathway, is expressed at lower levels in the lungs of patients with IPF. We also show substantial induction of COX-2, PTGES, prostaglandin E receptor 4 (EP4), and cytosolic phospholipase A2 (cPLA2) expression in human lung fibroblasts cultured in soft collagen hydrogels or in spheroids compared with conventional culture on stiff plastic culture plates. Induction of COX-2, cPLA2, and PTGES expression in spheroid cultures was moderately inhibited by the p38 mitogen-activated protein kinase inhibitor SB203580. The induction of prostanoid biosynthetic enzyme expression was accompanied by an increase in PGE2 levels only in non-IPF-derived fibroblast spheroids. Our study reveals an extensive dysregulation of prostanoid biosynthesis and signaling pathways in IPF-derived fibroblasts, which are only partially abrogated by culture in soft microenvironments.
Collapse
Affiliation(s)
- Asres Berhan
- Department of Pharmacology and Therapeutics, and
| | - Trudi Harris
- Department of Pharmacology and Therapeutics, and
| | - Jade Jaffar
- Department of Allergy, Immunology, Respiratory Medicine, The Alfred Hospital/Monash University, Melbourne, Victoria, Australia
| | - Fernando Jativa
- Department of Pharmacology and Therapeutics, and.,Department of Biomedical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | - Milica Ng
- CSL Ltd., Melbourne, Victoria, Australia; and
| | - Peter Lee
- Department of Biomedical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Glen Westall
- Department of Allergy, Immunology, Respiratory Medicine, The Alfred Hospital/Monash University, Melbourne, Victoria, Australia
| | - Nick Wilson
- CSL Ltd., Melbourne, Victoria, Australia; and
| | | | - Alastair G Stewart
- Department of Pharmacology and Therapeutics, and.,ARC Centre for Personalised Therapeutics Technologies, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Wang J, Zhao X, Feng W, Li Y, Peng C. Inhibiting TGF-[Formula: see text] 1-Mediated Cellular Processes as an Effective Strategy for the Treatment of Pulmonary Fibrosis with Chinese Herbal Medicines. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1965-1999. [PMID: 34961416 DOI: 10.1142/s0192415x21500932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pulmonary fibrosis (PF) is a chronic and irreversible interstitial lung disease that even threatens the lives of some patients infected with COVID-19. PF is a multicellular pathological process, including the initial injuries of epithelial cells, recruitment of inflammatory cells, epithelial-mesenchymal transition, activation and differentiation of fibroblasts, etc. TGF-[Formula: see text]1 acts as a key effect factor that participates in these cellular processes of PF. Recently, much attention was paid to inhibiting TGF-[Formula: see text]1 mediated cell processes in the treatment of PF with Chinese herbal medicines (CHM), an important part of traditional Chinese medicine. Here, this review first summarized the effects of TGF-[Formula: see text]1 in different cellular processes of PF. Then, this review summarized the recent research on CHM (compounds, multi-components, single medicines and prescriptions) to directly and/or indirectly inhibit TGF-[Formula: see text]1 signaling (TLRs, PPARs, micrRNA, etc.) in PF. Most of the research focused on CHM natural compounds, including but not limited to alkaloids, flavonoids, phenols and terpenes. After review, the research perspectives of CHM on TGF-[Formula: see text]1 inhibition in PF were further discussed. This review hopes that revealing the inhibiting effects of CHM on TGF-[Formula: see text]1-mediated cellular processes of PF can promote CHM to be better understood and utilized, thus transforming the therapeutic activities of CHM into practice.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Xingtao Zhao
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Wuwen Feng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Yunxia Li
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Cheng Peng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| |
Collapse
|
33
|
Matsuda S, Kim JD, Sugiyama F, Matsuo Y, Ishida J, Murata K, Nakamura K, Namiki K, Sudo T, Kuwaki T, Hatano M, Tatsumi K, Fukamizu A, Kasuya Y. Transcriptomic Evaluation of Pulmonary Fibrosis-Related Genes: Utilization of Transgenic Mice with Modifying p38 Signal in the Lungs. Int J Mol Sci 2020; 21:E6746. [PMID: 32937976 PMCID: PMC7555042 DOI: 10.3390/ijms21186746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 12/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing lung disease that is caused by the dysregulation of alveolar epithelial type II cells (AEC II). The mechanisms involved in the progression of IPF remain incompletely understood, although the immune response accompanied by p38 mitogen-activated protein kinase (MAPK) activation may contribute to some of them. This study aimed to examine the association of p38 activity in the lungs with bleomycin (BLM)-induced pulmonary fibrosis and its transcriptomic profiling. Accordingly, we evaluated BLM-induced pulmonary fibrosis during an active fibrosis phase in three genotypes of mice carrying stepwise variations in intrinsic p38 activity in the AEC II and performed RNA sequencing of their lungs. Stepwise elevation of p38 signaling in the lungs of the three genotypes was correlated with increased severity of BLM-induced pulmonary fibrosis exhibiting reduced static compliance and higher collagen content. Transcriptome analysis of these lung samples also showed that the enhanced p38 signaling in the lungs was associated with increased transcription of the genes driving the p38 MAPK pathway and differentially expressed genes elicited by BLM, including those related to fibrosis as well as the immune system. Our findings underscore the significance of p38 MAPK in the progression of pulmonary fibrosis.
Collapse
Affiliation(s)
- Shuichi Matsuda
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba City, Chiba 260-8670, Japan; (S.M.); (M.H.)
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba City, Chiba 260-8670, Japan; (Y.M.); (K.T.)
| | - Jun-Dal Kim
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan; (J.-D.K.); (J.I.); (K.M.); (A.F.)
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center and Trans-Border Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan;
| | - Yuji Matsuo
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba City, Chiba 260-8670, Japan; (Y.M.); (K.T.)
| | - Junji Ishida
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan; (J.-D.K.); (J.I.); (K.M.); (A.F.)
| | - Kazuya Murata
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan; (J.-D.K.); (J.I.); (K.M.); (A.F.)
- Laboratory Animal Resource Center and Trans-Border Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan;
| | - Kanako Nakamura
- Graduate School of Sciences and Technology, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan;
| | - Kana Namiki
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, Chiba City, Chiba 260-8670, Japan;
| | - Tatsuhiko Sudo
- Chemical Biology Core Facility and Antibiotics Laboratory, RIKEN Advanced Science Institute, Wako, Saitama 351-0198, Japan;
| | - Tomoyuki Kuwaki
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City, Kagoshima 890-8544, Japan;
| | - Masahiko Hatano
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba City, Chiba 260-8670, Japan; (S.M.); (M.H.)
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba City, Chiba 260-8670, Japan; (Y.M.); (K.T.)
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan; (J.-D.K.); (J.I.); (K.M.); (A.F.)
| | - Yoshitoshi Kasuya
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba City, Chiba 260-8670, Japan; (S.M.); (M.H.)
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, Chiba City, Chiba 260-8670, Japan;
| |
Collapse
|
34
|
Aschner Y, Nelson M, Brenner M, Roybal H, Beke K, Meador C, Foster D, Correll KA, Reynolds PR, Anderson K, Redente EF, Matsuda J, Riches DWH, Groshong SD, Pozzi A, Sap J, Wang Q, Rajshankar D, McCulloch CAG, Zemans RL, Downey GP. Protein tyrosine phosphatase-α amplifies transforming growth factor-β-dependent profibrotic signaling in lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2020; 319:L294-L311. [PMID: 32491951 PMCID: PMC7473933 DOI: 10.1152/ajplung.00235.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 04/06/2020] [Accepted: 04/25/2020] [Indexed: 01/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, often fatal, fibrosing lung disease for which treatment remains suboptimal. Fibrogenic cytokines, including transforming growth factor-β (TGF-β), are central to its pathogenesis. Protein tyrosine phosphatase-α (PTPα) has emerged as a key regulator of fibrogenic signaling in fibroblasts. We have reported that mice globally deficient in PTPα (Ptpra-/-) were protected from experimental pulmonary fibrosis, in part via alterations in TGF-β signaling. The goal of this study was to determine the lung cell types and mechanisms by which PTPα controls fibrogenic pathways and whether these pathways are relevant to human disease. Immunohistochemical analysis of lungs from patients with IPF revealed that PTPα was highly expressed by mesenchymal cells in fibroblastic foci and by airway and alveolar epithelial cells. To determine whether PTPα promotes profibrotic signaling pathways in lung fibroblasts and/or epithelial cells, we generated mice with conditional (floxed) Ptpra alleles (Ptpraf/f). These mice were crossed with Dermo1-Cre or with Sftpc-CreERT2 mice to delete Ptpra in mesenchymal cells and alveolar type II cells, respectively. Dermo1-Cre/Ptpraf/f mice were protected from bleomycin-induced pulmonary fibrosis, whereas Sftpc-CreERT2/Ptpraf/f mice developed pulmonary fibrosis equivalent to controls. Both canonical and noncanonical TGF-β signaling and downstream TGF-β-induced fibrogenic responses were attenuated in isolated Ptpra-/- compared with wild-type fibroblasts. Furthermore, TGF-β-induced tyrosine phosphorylation of TGF-β type II receptor and of PTPα were attenuated in Ptpra-/- compared with wild-type fibroblasts. The phenotype of cells genetically deficient in PTPα was recapitulated with the use of a Src inhibitor. These findings suggest that PTPα amplifies profibrotic TGF-β-dependent pathway signaling in lung fibroblasts.
Collapse
Affiliation(s)
- Yael Aschner
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Meghan Nelson
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Matthew Brenner
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Helen Roybal
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Keriann Beke
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Carly Meador
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Daniel Foster
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Kelly A Correll
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Paul R Reynolds
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Kelsey Anderson
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado
| | - Elizabeth F Redente
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado
- Veterans Affairs Eastern Colorado Heath Care System, Denver, Colorado
| | - Jennifer Matsuda
- Department of Biomedical Research, National Jewish Health, Denver, Colorado
| | - David W H Riches
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado
- Veterans Affairs Eastern Colorado Heath Care System, Denver, Colorado
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado
| | - Steve D Groshong
- Division of Pathology, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Ambra Pozzi
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Veterans Affairs Medical Center, Nashville, Tennessee
| | - Jan Sap
- Epigenetics and Cell Fate, Université Paris, Paris, France
| | - Qin Wang
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Dhaarmini Rajshankar
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | | | - Rachel L Zemans
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
- Department of Pediatrics, National Jewish Health, Denver, Colorado
- Department of Biomedical Research, National Jewish Health, Denver, Colorado
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado
| |
Collapse
|
35
|
Conforti F, Ridley R, Brereton C, Alzetani A, Johnson B, Marshall BG, Fletcher SV, Ottensmeier CH, Richeldi L, Skipp P, Wang Y, Jones MG, Davies DE. Paracrine SPARC signaling dysregulates alveolar epithelial barrier integrity and function in lung fibrosis. Cell Death Discov 2020; 6:54. [PMID: 32637156 PMCID: PMC7327077 DOI: 10.1038/s41420-020-0289-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/14/2020] [Accepted: 06/08/2020] [Indexed: 12/23/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic scarring disease in which aging, environmental exposure(s) and genetic susceptibility have been implicated in disease pathogenesis, however, the causes and mechanisms of the progressive fibrotic cascade are still poorly understood. As epithelial-mesenchymal interactions are essential for normal wound healing, through human 2D and 3D in vitro studies, we tested the hypothesis that IPF fibroblasts (IPFFs) dysregulate alveolar epithelial homeostasis. Conditioned media from IPFFs exaggerated the wound-healing response of primary human Type II alveolar epithelial cells (AECs). Furthermore, AECs co-cultured with IPFFs exhibited irregular epithelialization compared with those co-cultured with control fibroblasts (NHLFs) or AECs alone, suggesting that epithelial homeostasis is dysregulated in IPF as a consequence of the abnormal secretory phenotype of IPFFs. Secretome analysis of IPFF conditioned media and functional studies identified the matricellular protein, SPARC, as a key mediator in the epithelial-mesenchymal paracrine signaling, with increased secretion of SPARC by IPFFs promoting persistent activation of alveolar epithelium via an integrin/focal adhesion/cellular-junction axis resulting in disruption of epithelial barrier integrity and increased macromolecular permeability. These findings suggest that in IPF fibroblast paracrine signaling promotes persistent alveolar epithelial activation, so preventing normal epithelial repair responses and restoration of tissue homeostasis. Furthermore, they identify SPARC-mediated paracrine signaling as a potential therapeutic target to promote the restoration of lung epithelial homoestasis in IPF patients.
Collapse
Affiliation(s)
- Franco Conforti
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
| | - Robert Ridley
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - Christopher Brereton
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
| | - Aiman Alzetani
- Department of Thoracic Surgery, University Hospital Southampton, Southampton, SO16 6YD UK
- University Hospital Southampton, Southampton, SO16 6YD UK
| | - Benjamin Johnson
- Cancer Sciences & NIHR and CRUK Experimental Cancer Sciences Unit, University of Southampton, Southampton, SO16 6YD UK
| | - Ben G. Marshall
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- University Hospital Southampton, Southampton, SO16 6YD UK
| | - Sophie V. Fletcher
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- University Hospital Southampton, Southampton, SO16 6YD UK
| | - Christian H. Ottensmeier
- University Hospital Southampton, Southampton, SO16 6YD UK
- Cancer Sciences & NIHR and CRUK Experimental Cancer Sciences Unit, University of Southampton, Southampton, SO16 6YD UK
| | - Luca Richeldi
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- Unità Operativa Complessa di Pneumologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Paul Skipp
- Centre for Proteomic Research, Institute for Life Sciences University of Southampton, Southampton, SO17 1BJ UK
| | - Yihua Wang
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ UK
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, SO17 1BJ UK
| | - Mark G. Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- University Hospital Southampton, Southampton, SO16 6YD UK
| | - Donna E. Davies
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ UK
| |
Collapse
|
36
|
Wang A, Cao S, Aboelkassem Y, Valdez-Jasso D. Quantification of uncertainty in a new network model of pulmonary arterial adventitial fibroblast pro-fibrotic signalling. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2020; 378:20190338. [PMID: 32448066 PMCID: PMC7287331 DOI: 10.1098/rsta.2019.0338] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/16/2020] [Indexed: 05/21/2023]
Abstract
Here, we present a novel network model of the pulmonary arterial adventitial fibroblast (PAAF) that represents seven signalling pathways, confirmed to be important in pulmonary arterial fibrosis, as 92 reactions and 64 state variables. Without optimizing parameters, the model correctly predicted 80% of 39 results of input-output and inhibition experiments reported in 20 independent papers not used to formulate the original network. Parameter uncertainty quantification (UQ) showed that this measure of model accuracy is robust to changes in input weights and half-maximal activation levels (EC50), but is more affected by uncertainty in the Hill coefficient (n), which governs the biochemical cooperativity or steepness of the sigmoidal activation function of each state variable. Epistemic uncertainty in model structure, due to the reliance of some network components and interactions on experiments using non-PAAF cell types, suggested that this source of uncertainty had a smaller impact on model accuracy than the alternative of reducing the network to only those interactions reported in PAAFs. UQ highlighted model parameters that can be optimized to improve prediction accuracy and network modules where there is the greatest need for new experiments. This article is part of the theme issue 'Uncertainty quantification in cardiac and cardiovascular modelling and simulation'.
Collapse
Affiliation(s)
| | | | | | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92092, USA
| |
Collapse
|
37
|
Tan Y, Qiao Y, Chen Z, Liu J, Guo Y, Tran T, Tan KS, Wang DY, Yan Y. FGF2, an Immunomodulatory Factor in Asthma and Chronic Obstructive Pulmonary Disease (COPD). Front Cell Dev Biol 2020; 8:223. [PMID: 32300593 PMCID: PMC7142218 DOI: 10.3389/fcell.2020.00223] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
The fibroblast growth factor 2 (FGF2) is a potent mitogenic factor belonging to the FGF family. It plays a role in airway remodeling associated with chronic inflammatory airway diseases, including asthma and chronic obstructive pulmonary disease (COPD). Recently, research interest has been raised in the immunomodulatory function of FGF2 in asthma and COPD, through its involvement in not only the regulation of inflammatory cells but also its participation as a mediator between immune cells and airway structural cells. Herein, this review provides the current knowledge on the biology of FGF2, its expression pattern in asthma and COPD patients, and its role as an immunomodulatory factor. The potential that FGF2 is involved in regulating inflammation indicates that FGF2 could be a therapeutic target for chronic inflammatory diseases.
Collapse
Affiliation(s)
- Yuanyang Tan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | | | - Zhuanggui Chen
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Liu
- Department of Respiratory Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yanrong Guo
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai Sen Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, University Health System, National University of Singapore, Singapore, Singapore
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, University Health System, National University of Singapore, Singapore, Singapore
| | - Yan Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
38
|
Goda C, Balli D, Black M, Milewski D, Le T, Ustiyan V, Ren X, Kalinichenko VV, Kalin TV. Loss of FOXM1 in macrophages promotes pulmonary fibrosis by activating p38 MAPK signaling pathway. PLoS Genet 2020; 16:e1008692. [PMID: 32271749 PMCID: PMC7173935 DOI: 10.1371/journal.pgen.1008692] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 04/21/2020] [Accepted: 02/22/2020] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic disease with high mortality and is refractory to treatment. Pulmonary macrophages can both promote and repress fibrosis, however molecular mechanisms regulating macrophage functions during fibrosis remain poorly understood. FOXM1 is a transcription factor and is not expressed in quiescent lungs. Herein, we show that FOXM1 is highly expressed in pulmonary macrophages within fibrotic lungs of IPF patients and mouse fibrotic lungs. Macrophage-specific deletion of Foxm1 in mice (myFoxm1-/-) exacerbated pulmonary fibrosis. Inactivation of FOXM1 in vivo and in vitro increased p38 MAPK signaling in macrophages and decreased DUSP1, a negative regulator of p38 MAPK pathway. FOXM1 directly activated Dusp1 promoter. Overexpression of DUSP1 in FOXM1-deficient macrophages prevented activation of p38 MAPK pathway. Adoptive transfer of wild-type monocytes to myFoxm1-/- mice alleviated bleomycin-induced fibrosis. Altogether, contrary to known pro-fibrotic activities in lung epithelium and fibroblasts, FOXM1 has anti-fibrotic function in macrophages by regulating p38 MAPK.
Collapse
Affiliation(s)
- Chinmayee Goda
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - David Balli
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Markaisa Black
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - David Milewski
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Tien Le
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Vladimir Ustiyan
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
- Center for Lung Regenerative Medicine, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Xiaomeng Ren
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
- Center for Lung Regenerative Medicine, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Vladimir V. Kalinichenko
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
- Center for Lung Regenerative Medicine, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Tanya V. Kalin
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| |
Collapse
|
39
|
Janulaityte I, Januskevicius A, Kalinauskaite-Zukauske V, Bajoriuniene I, Malakauskas K. In Vivo Allergen-Activated Eosinophils Promote Collagen I and Fibronectin Gene Expression in Airway Smooth Muscle Cells via TGF- β1 Signaling Pathway in Asthma. Int J Mol Sci 2020; 21:E1837. [PMID: 32155894 PMCID: PMC7084581 DOI: 10.3390/ijms21051837] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/01/2020] [Accepted: 03/05/2020] [Indexed: 12/30/2022] Open
Abstract
Eosinophils infiltration and releasing TGF-β1 in the airways has been implicated in the pathogenesis of asthma, especially during acute episodes provoked by an allergen. TGF-β1 is a major mediator involved in pro-inflammatory responses and fibrotic tissue remodeling in asthma. We aimed to evaluate the effect of in vivo allergen-activated eosinophils on the expression of COL1A1 and FN in ASM cells in asthma. A total of 12 allergic asthma patients and 11 healthy subjects were examined. All study subjects underwent bronchial challenge with D. pteronyssinus allergen. Eosinophils from peripheral blood were isolated before and 24 h after the bronchial allergen challenge using high-density centrifugation and magnetic separation. Individual co-cultures of blood eosinophils and immortalized human ASM cells were prepared. The TGF-β1 concentration in culture supernatants was analyzed using ELISA. Gene expression was analyzed using qRT-PCR. Eosinophils integrins were suppressed with linear RGDS peptide before co-culture with ASM cells. Results: The expression of TGF-β1 in asthmatic eosinophils significantly increased over non-activated asthmatic eosinophils after allergen challenge, p < 0.001. The TGF-β1 concentration in culture supernatants was significantly higher in samples with allergen-activated asthmatic eosinophils compared to baseline, p < 0.05. The effect of allergen-activated asthmatic eosinophils on the expression of TGF-β1, COL1A1, and FN in ASM cells was more significant compared to non-activated eosinophils, p < 0.05, however, no difference was found on WNT-5A expression. The incubation of allergen-activated asthmatic eosinophils with RGDS peptide was more effective compared to non-activated eosinophils as the gene expression in ASM cells was downregulated equally to the same level as healthy eosinophils.
Collapse
Affiliation(s)
- Ieva Janulaityte
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (K.M.)
| | - Andrius Januskevicius
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (K.M.)
| | | | - Ieva Bajoriuniene
- Department of Immunology and Allergology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania;
| | - Kestutis Malakauskas
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (K.M.)
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania;
| |
Collapse
|
40
|
Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med 2020; 217:e20190103. [PMID: 32997468 PMCID: PMC7062524 DOI: 10.1084/jem.20190103] [Citation(s) in RCA: 596] [Impact Index Per Article: 119.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 12/24/2019] [Indexed: 12/21/2022] Open
Abstract
TGF-β is extensively implicated in the pathogenesis of fibrosis. In fibrotic lesions, spatially restricted generation of bioactive TGF-β from latent stores requires the cooperation of proteases, integrins, and specialized extracellular matrix molecules. Although fibroblasts are major targets of TGF-β, some fibrogenic actions may reflect activation of other cell types, including macrophages, epithelial cells, and vascular cells. TGF-β–driven fibrosis is mediated through Smad-dependent or non-Smad pathways and is modulated by coreceptors and by interacting networks. This review discusses the role of TGF-β in fibrosis, highlighting mechanisms of TGF-β activation and signaling, the cellular targets of TGF-β actions, and the challenges of therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
41
|
Hoffmann-Vold AM, Weigt SS, Saggar R, Palchevskiy V, Volkmann ER, Liang LL, Ross D, Ardehali A, Lynch JP, Belperio JA. Endotype-phenotyping may predict a treatment response in progressive fibrosing interstitial lung disease. EBioMedicine 2019; 50:379-386. [PMID: 31732480 PMCID: PMC6921223 DOI: 10.1016/j.ebiom.2019.10.050] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Some interstitial lung disease (ILD) patients develop a progressive fibrosing-ILD phenotype (PF-ILD), with similar persistent lung function decline suggesting common molecular pathways involved. Nintedanib, a tyrosine kinase inhibitor targeting the PDGF, FGF, VEGF and M-CSF pathways, has shown comparable efficacy in idiopathic pulmonary fibrosis (IPF) and systemic sclerosis-associated ILD (SSc-ILD). We hypothesize that Nintedanib targeted molecular pathways will be augmented to a similar degree across PF-ILD regardless of aetiology. METHODS We collected explanted lung tissue at the time of lung transplantation from 130 PF-ILD patients (99 (76%) IPF, 14 (11%) SSc-ILD, 17 (13%) other PF-ILD), and wedge biopsies from 200 donor lungs and measured PDGF, FGF, VEGF and M-CSF concentrations by Luminex. FINDINGS The concentrations of PDGF-AA, PDGF-BB, FGF-2, VEGF and M-CSF were significantly increased in PF-ILD lungs compared to donor lungs (PDGF-AA 93·0 pg/ml [±97·2] vs. 37·5 pg/ml [±35·4], p < 0·001; PDGF-BB 102·5 pg/ml [±78·8] vs. 61·9 pg/ml [±47·0], p < 0·001; FGF-2 1442·4 pg/ml [±426·6] vs. 1201·7 pg/ml [±535·2], p = 0·009; VEGF 40·6 pg/ml [±20·1] vs. 24·9 pg/ml [±29·5], p < 0·001; and M-CSF 25526 pg/ml [±24,799] vs. 6120 pg/ml [±7245], p < 0·001). There were no significant differences in these growth factor/angiogenic molecules/cytokine concentrations when segregated by IPF, SSc-ILD and other PF-ILDs. INTERPRETATION Nintedanib specific targeted molecular pathways are augmented to a similar magnitude in all PF-ILD lung tissue as compared to controls, suggesting that Nintedanib treatment may be efficacious in PF-ILD regardless of aetiology. We speculate that clinical trials using Nintedanib for PF-ILD with or without IPF or SSc-ILD should show a similar relative reduction in FVC decline as seen in IPF and SSc-ILD (∼45-50%). FUNDING Health Grant P01-HL108793 (JAB), South-Eastern Norway Regional Health Authority Grant 2018072 (AMHV).
Collapse
Affiliation(s)
- Anna-Maria Hoffmann-Vold
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Pb 4950 Nydalen, 0424 Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Rikshospitalet, Pb 4950 Nydalen, 0424 Oslo, Norway
| | - S Samuel Weigt
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Rajan Saggar
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Vyacheslav Palchevskiy
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Elizabeth R Volkmann
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Lloyd L Liang
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - David Ross
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Abbas Ardehali
- Department of Surgery, UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Joseph P Lynch
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - John A Belperio
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA.
| |
Collapse
|
42
|
Zhang P, Liu J, Zong R. Triptolide protects against TGF-β1-induced pulmonary fibrosis by regulating FAK/calpain signaling. Exp Ther Med 2019; 18:4781-4789. [PMID: 31777561 PMCID: PMC6862707 DOI: 10.3892/etm.2019.8127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 09/04/2019] [Indexed: 11/17/2022] Open
Abstract
The present study aimed to investigate the mechanism of anti-proliferative, anti-inflammatory and anti-fibrotic effects of triptolide (TPL) on activated lung fibroblasts by regulating the focal adhesion kinase (FAK) and calpain signaling pathways. The HFL-1 human foetal lung fibroblast cell line was cultured in vitro and treated with 50 ng/ml transforming growth factor (TGF)-β1 for 48 h to establish the model of pulmonary fibrosis. Subsequently, the cells were divided into five groups, including a control, model, TPL, FAK inhibitor and calpeptin group. Subsequently, the proliferation of lung fibroblasts was detected using the Cell Counting Kit-8 assay. The concentration of interleukin (IL)-6 in the cell culture supernatant was examined by ELISA and the mRNA expression levels of collagen type I (ColI)α and ColIII in lung fibroblasts were quantified by reverse transcription-quantitative PCR. The protein levels of FAK, phosphorylated (p)-FAK, calpain 1 and calpain 2 were detected by western blot analysis. TGF-β1 induced the proliferation of lung fibroblasts, whereas TPL inhibited this proliferation in a dose-dependent manner. TPL also decreased the TGF-β1-induced production of IL-6 and reduced the upregulation of ColIα, ColIII, FAK, p-FAK, and inhibited the decrease of calpain 1 and calpain 2 induced by TGF-β1. In addition, the FAK inhibitor acted synergistically with TPL to decrease TGF-β1-induced production of IL-6 and attenuate TGF-β1-induced synthesis of ColIα and ColIII, while calpeptin had an antagonistic effect on the function of TPL. Furthermore, treatment with the FAK inhibitor and TPL markedly decreased the protein levels of FAK and p-FAK, and increased the protein expression of calpain 1 and calpain 2 in lung fibroblasts stimulated by TGF-β1 to a greater extent than TPL alone, while calpeptin had an antagonistic effect on the action of TPL. In conclusion, the present study indicated that TPL protected against TGF-β1-induced proliferation, inflammation and fibrosis by regulating the FAK and calpain signaling pathways.
Collapse
Affiliation(s)
- Pingheng Zhang
- Traditional Chinese Medicine Department, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Jian Liu
- Rheumatism Department, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Ruikai Zong
- Rheumatism Department, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
43
|
Kim MH, Jung SY, Song KH, Park JI, Ahn J, Kim EH, Park JK, Hwang SG, Woo HJ, Song JY. A new FGFR inhibitor disrupts the TGF-β1-induced fibrotic process. J Cell Mol Med 2019; 24:830-840. [PMID: 31692229 PMCID: PMC6933341 DOI: 10.1111/jcmm.14793] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/11/2019] [Accepted: 09/23/2019] [Indexed: 01/06/2023] Open
Abstract
Pulmonary fibrosis (PF) is chronic and irreversible damage to the lung characterized by fibroblast activation and matrix deposition. Although recently approved novel anti‐fibrotic agents can improve the lung function and survival of patients with PF, the overall outcomes remain poor. In this study, a novel imidazopurine compound, 3‐(2‐chloro‐6‐fluorobenzyl)‐1,6,7‐trimethyl‐1H‐imidazo[2,1‐f]purine‐2,4(3H,8H)‐dione (IM‐1918), markedly inhibited transforming growth factor (TGF)‐β‐stimulated reporter activity and reduced the expression of representative fibrotic markers, such as connective tissue growth factor, fibronectin, collagen and α‐smooth muscle actin, on human lung fibroblasts. However, IM‐1918 neither decreased Smad‐2 and Smad‐3 nor affected p38MAPK and JNK. Instead, IM‐1918 reduced Akt and extracellular signal‐regulated kinase 1/2 phosphorylation increased by TGF‐β. Additionally, IM‐1918 inhibited the phosphorylation of fibroblast growth factor receptors 1 and 3. In a bleomycin‐induced murine lung fibrosis model, IM‐1918 profoundly reduced fibrotic areas and decreased collagen and α‐smooth muscle actin accumulation. These results suggest that IM‐1918 can be applied to treat lung fibrosis.
Collapse
Affiliation(s)
- Mi-Hyoung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea.,Laboratory of Immunology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Seung-Youn Jung
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Kyung-Hee Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Jeong-In Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Jiyeon Ahn
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Eun-Ho Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Jong Kuk Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Sang-Gu Hwang
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Hee-Jong Woo
- Laboratory of Immunology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Jie-Young Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| |
Collapse
|
44
|
Lee HS, Hua HS, Wang CH, Yu MC, Chen BC, Lin CH. Mycobacterium tuberculosis induces connective tissue growth factor expression through the TLR2-JNK-AP-1 pathway in human lung fibroblasts. FASEB J 2019; 33:12554-12564. [PMID: 31451010 DOI: 10.1096/fj.201900487r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mycobacterium tuberculosis (M.tb) infection in lung causes pulmonary fibrosis, which leads to the irreversible reduction of pulmonary function. Fibrotic protein connective tissue growth factor (CTGF) expression has been confirmed to play a crucial role in lung fibrosis. However, the underlying signal pathway and effect of M.tb on CTGF expression in human lung fibroblasts are unclear. Our results revaled that M.tb caused time- and concentration-dependent increases in CTGF expression in human lung fibroblasts. A mechanistic investigation revealed that M.tb induced CTGF expression through TLR2 but not TLR4. The promoter activity assay indicated that M.tb-induced CTGF activity was mainly controlled by the promoter region at -747 to -184 bp, which contained signal transducer and activator of transcription 3 and activator protein 1 (AP-1) binding sites. Moreover, curcumin (AP-1 inhibitor) restrained M.tb-induced CTGF expression. M.tb also induced increases in AP-1 luciferase activity and DNA binding activity of c-Jun and c-Fos on the CTGF promoter. Furthermore, the knockdown of c-Jun by small interfering RNA attenuated M.tb-induced CTGF expression and AP-1 luciferase activity. A JNK inhibitor (SP600125) and a JNK dominant-negative mutant suppressed M.tb-induced CTGF expression. We also discovered that M.tb could induce the phosphorylation of JNK and c-Jun. Furthermore, SP600125 inhibited M.tb-induced c-Jun phosphorylation and AP-1- luciferase activity. M.tb-induced fibronectin expression was inhibited by anti-CTGF antibody. These results demonstrate that M.tb is activated through TLR2 to induce JNK activation, further increasing the DNA binding activity of c-Jun and c-Fos and finally inducing CTGF expression and extracellular matrix production.-Lee, H.-S., Hua, H.-S., Wang, C.-H., Yu, M.-C., Chen, B.-C., Lin, C.-H. Mycobacterium tuberculosis induces connective tissue growth factor expression through the TLR2-JNK-AP-1 pathway in human lung fibroblasts.
Collapse
Affiliation(s)
- Hong-Sheng Lee
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Hung-Sheng Hua
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Hui Wang
- Department of Laboratory Medicine, Wanfang Hospital, Taipei Medical University, Taipei, Taiwan.,Pulmonary Research Center, Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ming-Chih Yu
- Pulmonary Research Center, Wanfang Hospital, Taipei Medical University, Taipei, Taiwan.,School of Respiratory Therapy, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Bing-Chang Chen
- School of Respiratory Therapy, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Huang Lin
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
45
|
Laddha AP, Kulkarni YA. VEGF and FGF-2: Promising targets for the treatment of respiratory disorders. Respir Med 2019; 156:33-46. [PMID: 31421589 DOI: 10.1016/j.rmed.2019.08.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/19/2022]
Abstract
The endothelial cells play a crucial role in the progression of angiogenesis, which causes cell re-modulation, proliferation, adhesion, migration, invasion and survival. Angiogenic factors like cytokines, cell adhesion molecules, growth factors, vasoactive peptides, proteolytic enzymes (metalloproteinases) and plasminogen activators bind to their receptors on endothelial cells and activate the signal transduction pathways like epidermal growth factor receptor (EGFR phosphatidylinositol 3-kinase and (PI3K)/AKT/mammalian target of rapamycin (mTOR) which initiate the process of angiogenesis. Cytokines that stimulate angiogenesis include direct and indirect proangiogenic markers. The direct proangiogenic group of markers consists of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (FGF-2) and hepatocyte growth factor (HGF) whereas the indirect proangiogenic markers include transforming growth factor-beta (TGF-β), interleukin 6 (IL-6), interleukin 8 (IL-8) and platelet-derived growth factor (PDGF). VEGF and FGF-2 are the strongest activators of angiogenesis which stimulate migration and proliferation of endothelial cells in existing vessels to generate and stabilize new blood vessels. VEGF is released in hypoxic conditions as an effect of the hypoxia-inducible factor (HIF-1α) and causes re-modulation and inflammation of bronchi cell. Cell re-modulation and inflammation leads to the development of various lung disorders like pulmonary hypertension, chronic obstructive pulmonary disease, asthma, fibrosis and lung cancer. This indicates that there is a firm link between overexpression of VEGF and FGF-2 with lung disorders. Various natural and synthetic drugs are available for reducing the overexpression of VEGF and FGF-2 which can be helpful in treating lung disorders. Researchers are still searching for new angiogenic inhibitors which can be helpful in the treatment of lung disorders. The present review emphasizes on molecular mechanisms and new drug discovery focused on VEGF and FGF-2 inhibitors and their role as anti-angiogenetic agents in lung disorders.
Collapse
Affiliation(s)
- Ankit P Laddha
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400 056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400 056, India.
| |
Collapse
|
46
|
Ballester B, Milara J, Cortijo J. Idiopathic Pulmonary Fibrosis and Lung Cancer: Mechanisms and Molecular Targets. Int J Mol Sci 2019; 20:ijms20030593. [PMID: 30704051 PMCID: PMC6387034 DOI: 10.3390/ijms20030593] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/18/2019] [Accepted: 01/28/2019] [Indexed: 12/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common idiopathic interstitial pulmonary disease with a median survival of 2–4 years after diagnosis. A significant number of IPF patients have risk factors, such as a history of smoking or concomitant emphysema, both of which can predispose the patient to lung cancer (LC) (mostly non-small cell lung cancer (NSCLC)). In fact, IPF itself increases the risk of LC development by 7% to 20%. In this regard, there are multiple common genetic, molecular, and cellular processes that connect lung fibrosis with LC, such as myofibroblast/mesenchymal transition, myofibroblast activation and uncontrolled proliferation, endoplasmic reticulum stress, alterations of growth factors expression, oxidative stress, and large genetic and epigenetic variations that can predispose the patient to develop IPF and LC. The current approved IPF therapies, pirfenidone and nintedanib, are also active in LC. In fact, nintedanib is approved as a second line treatment in NSCLC, and pirfenidone has shown anti-neoplastic effects in preclinical studies. In this review, we focus on the current knowledge on the mechanisms implicated in the development of LC in patients with IPF as well as in current IPF and LC-IPF candidate therapies based on novel molecular advances.
Collapse
Affiliation(s)
- Beatriz Ballester
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBERES, Health Institute Carlos III, 28029 Valencia, Spain.
| | - Javier Milara
- CIBERES, Health Institute Carlos III, 28029 Valencia, Spain.
- Pharmacy Unit, University Clinic Hospital of Valencia, 46010 Valencia, Spain.
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain.
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBERES, Health Institute Carlos III, 28029 Valencia, Spain.
- Research and teaching Unit, University General Hospital Consortium, 46014 Valencia, Spain.
| |
Collapse
|
47
|
Jin YK, Li XH, Wang W, Liu J, Zhang W, Fang YS, Zhang ZF, Dai HP, Ning W, Wang C. Follistatin-Like 1 Promotes Bleomycin-Induced Pulmonary Fibrosis through the Transforming Growth Factor Beta 1/Mitogen-Activated Protein Kinase Signaling Pathway. Chin Med J (Engl) 2018; 131:1917-1925. [PMID: 30082522 PMCID: PMC6085847 DOI: 10.4103/0366-6999.238151] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background Follistatin-like 1 (FSTL1) is a novel profibrogenic factor that induces pulmonary fibrosis (PF) through the transforming growth factor-beta 1 (TGF-β1)/Smad signaling. Little is known about its effects on PF through the non-Smad signaling, like the mitogen-activated protein kinase (MAPK) pathway. Therefore, this study aimed to investigate the role of FSTL1 in PF through the MAPK signaling pathway and its mechanisms in lung fibrogenesis. Methods PF was induced in Fstl1+/-and wild-type (WT) C57BL/6 mice with bleomycin. After 14 days, the mice were sacrificed, and lung tissues were stained with hematoxylin and eosin; the hydroxyproline content was measured to confirm PF. The mRNA and protein level of FSTL1 and the change of MAPK phosphorylation were measured by quantitative polymerase chain reaction and Western blotting. The effect of Fstl1 deficiency on fibroblasts differentiation was measured by Western blotting and cell immunofluorescence. MAPK signaling activation was measured by Western blotting in Fstl1+/- and WT fibroblasts treated with recombinant human FSTL1 protein. We pretreated mouse lung fibroblast cells with inhibitors of the extracellular signal-regulated kinase (ERK), p38, and Jun N-terminal kinase (JNK) signaling and analyzed their differentiation, proliferation, migration, and invasion by Western blotting, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analysis, and transwell assays. The Student's t-test was used to compare the differences between two groups. Results Fstl1 deficiency attenuated phosphorylation of the ERK, p38, and JNK signaling in bleomycin-induced fibrotic lung tissue 14 days after injury (0.67 ± 0.05 vs. 1.22 ± 0.03, t = 14.92, P = 0.0001; 0.41 ± 0.01 vs. 1.15 ± 0.07; t = 11.19; P = 0.0004; and 0.41 ± 0.01 vs. 1.07 ± 0.07, t = 8.92, P = 0.0009; respectively), compared with WT lungs at the same time and in primary lung fibroblasts (0.82 ± 0.01 vs. 1.01 ± 0.04, t = 4.06, P = 0.0150; 1.04 ± 0.03 vs. 1.24 ± 0.03, t = 4.44, P = 0.0100; and 0.76 ± 0.05 vs. 0.99 ± 0.05, t = 4.48, P = 0.0100; respectively), compared with TGF-β1-stimulated WT group. Recombinant human FSTL1 protein in lung fibroblasts enhanced TGF-β1-mediated phosphorylation of the ERK (1.19 ± 0.08 vs. 0.55 ± 0.04, t = 6.99, P = 0.0020), p38 (1.18 ± 0.04 vs. 0.66 ± 0.03, t = 11.20, P = 0.0020), and JNK (1.11 ± 0.01 vs. 0.84 ± 0.04, t = 6.53, P = 0.0030), compared with the TGF-β1-stimulated WT group. Fstl1-deficient fibroblasts showed reduced alpha-smooth muscle actin (α-SMA) expression (0.70 ± 0.06 vs. 1.28 ± 0.11, t = 4.65, P = 0.0035, compared with the untreated WT group; 1.40 ± 0.05 vs. 1.76 ± 0.02, t = 6.31, P = 0.0007; compared with the TGF-β1-treated WT group). Compared with the corresponding condition in the control group, the TGF-β1/FSTL1-mediated α-SMA expression was significantly suppressed by pretreatment with an inhibitor of p38 (0.73 ± 0.01 vs. 1.13 ± 0.10, t = 3.92, P = 0.0078) and JNK (0.78 ± 0.03 vs. 1.08 ± 0.06, t = 4.40, P = 0.0046) signaling. The proliferation of mouse lung fibroblast cells (MLgs) significantly decreased after treatment of an inhibitor of p38 (0.30 ± 0.01 vs. 0.46 ± 0.03, t = 4.64, P = 0.0009), JNK (0.30 ± 0.01 vs. 0.49 ± 0.01, t = 12.84, P = 0.0001), and Smad2/3 (0.18 ± 0.02 vs. 0.46 ± 0.02, t = 12.69, P = 0.0001) signaling compared with the dimethylsulfoxide group. The migration and invasion cells of MLgs significantly decreased in medium pretreated with an inhibitor of p38 (70.17 ± 3.28 vs. 116.30 ± 7.11, t = 5.89, P = 0.0042 for the migratory cells; 19.87 ± 0.84 vs. 32.70 ± 0.95, t = 10.14, P = 0.0005 for the invasive cells), JNK (72.30 ± 3.85 vs. 116.30 ± 7.11, t = 5.44, P = 0.0056 for the migratory cells; 18.03 ± 0.94 vs. 32.70 ± 0.95, t = 11.00, P = 0.0004 for the invasive cells), and Smad2/3 (64.76 ± 1.41 vs. 116.30 ± 7.11, t = 7.11, P = 0.0021 for the migratory cells; 18.03 ± 0.94 vs. 32.70 ± 0.95, t = 13.29, P = 0.0002 for the invasive cells) signaling compared with the corresponding condition in the dimethylsulfoxide group. Conclusion FSTL1 affects lung fibroblast differentiation, proliferation, migration, and invasion through p38 and JNK signaling, and in this way, it might influence the development of PF.
Collapse
Affiliation(s)
- Yan-Kun Jin
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University; Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Respiratory Disease, Capital Medical University, Beijing 100020, China
| | - Xiao-He Li
- Department of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Wang Wang
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | - Jie Liu
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | - Wei Zhang
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | - Yin-Shan Fang
- Department of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhi-Fei Zhang
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | - Hua-Ping Dai
- Department of Respiratory Disease, Capital Medical University, Beijing 100020, China
| | - Wen Ning
- Department of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chen Wang
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Respiratory Disease, Capital Medical University, Beijing 100020, China
| |
Collapse
|
48
|
Michalik M, Wójcik-Pszczoła K, Paw M, Wnuk D, Koczurkiewicz P, Sanak M, Pękala E, Madeja Z. Fibroblast-to-myofibroblast transition in bronchial asthma. Cell Mol Life Sci 2018; 75:3943-3961. [PMID: 30101406 PMCID: PMC6182337 DOI: 10.1007/s00018-018-2899-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 07/26/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022]
Abstract
Bronchial asthma is a chronic inflammatory disease in which bronchial wall remodelling plays a significant role. This phenomenon is related to enhanced proliferation of airway smooth muscle cells, elevated extracellular matrix protein secretion and an increased number of myofibroblasts. Phenotypic fibroblast-to-myofibroblast transition represents one of the primary mechanisms by which myofibroblasts arise in fibrotic lung tissue. Fibroblast-to-myofibroblast transition requires a combination of several types of factors, the most important of which are divided into humoural and mechanical factors, as well as certain extracellular matrix proteins. Despite intensive research on the nature of this process, its underlying mechanisms during bronchial airway wall remodelling in asthma are not yet fully clarified. This review focuses on what is known about the nature of fibroblast-to-myofibroblast transition in asthma. We aim to consider possible mechanisms and conditions that may play an important role in fibroblast-to-myofibroblast transition but have not yet been discussed in this context. Recent studies have shown that some inherent and previously undescribed features of fibroblasts can also play a significant role in fibroblast-to-myofibroblast transition. Differences observed between asthmatic and non-asthmatic bronchial fibroblasts (e.g., response to transforming growth factor β, cell shape, elasticity, and protein expression profile) may have a crucial influence on this phenomenon. An accurate understanding and recognition of all factors affecting fibroblast-to-myofibroblast transition might provide an opportunity to discover efficient methods of counteracting this phenomenon.
Collapse
Affiliation(s)
- Marta Michalik
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Wójcik-Pszczoła
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| | - Milena Paw
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Dawid Wnuk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Paulina Koczurkiewicz
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Marek Sanak
- Division of Molecular Biology and Clinical Genetics, Department of Medicine, Jagiellonian University Medical College, Skawińska 8, 31-066, Kraków, Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| |
Collapse
|
49
|
Pollard BS, Pollard HB. Induced pluripotent stem cells for treating cystic fibrosis: State of the science. Pediatr Pulmonol 2018; 53:S12-S29. [PMID: 30062693 DOI: 10.1002/ppul.24118] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/31/2018] [Indexed: 12/20/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are a recently developed technology in which fully differentiated cells such as fibroblasts from individual CF patients can be repaired with [wildtype] CFTR, and reprogrammed to differentiate into fully differentiated cells characteristic of the proximal and distal airways. Here, we review properties of different epithelial cells in the airway, and the in vitro genetic roadmap which iPSCs follow as they are step-wise differentiated into either basal stem cells, for the proximal airway, or into Type II Alveolar cells for the distal airways. The central theme is that iPSC-derived basal stem cells, are penultimately dependent on NOTCH signaling for differentiation into club cells, goblet cells, ciliated cells, and neuroendocrine cells. Furthermore, given the proper matrix, these cellular progenies are also able to self-assemble into a fully functional pseudostratified squamous proximal airway epithelium. By contrast, club cells are reserve stem cells which are able to either differentiate into goblet or ciliated cells, but also to de-differentiate into basal stem cells. Variant club cells, located at the transition between airway and alveoli, may also be responsible for differentiation into Type II Alveolar cells, which then differentiate into Type I Alveolar cells for gas exchange in the distal airway. Using gene editing, the mutant CFTR gene in iPSCs from CF patients can be repaired, and fully functional epithelial cells can thus be generated through directed differentiation. However, there is a limitation in that the lung has other CFTR-dependent cells besides epithelial cells. Another limitation is that there are CFTR-dependent cells in other organs which would continue to contribute to CF disease. Furthermore, there are also bystander or modifier genes which affect disease outcome, not only in the lung, but specifically in other CF-affected organs. Finally, we discuss future personalized applications of the iPSC technology, many of which have already survived the "proof-of-principle" test. These include (i) patient-derived iPSCs used as a "lung-on-a-chip" tool for personalized drug discovery; (ii) replacement of mutant lung cells by wildtype lung cells in the living lung; and (iii) development of bio-artificial lungs. It is hoped that this review will give the reader a roadmap through the most complicated of the obstacles, and foster a guardedly optimistic view of how some of the remaining obstacles might one day be overcome.
Collapse
Affiliation(s)
| | - Harvey B Pollard
- Department of Cell Biology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
50
|
Hewlett JC, Kropski JA, Blackwell TS. Idiopathic pulmonary fibrosis: Epithelial-mesenchymal interactions and emerging therapeutic targets. Matrix Biol 2018; 71-72:112-127. [PMID: 29625182 PMCID: PMC6146058 DOI: 10.1016/j.matbio.2018.03.021] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/11/2018] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic fibrotic disease of the lung that is marked by progressive decline in pulmonary function and ultimately respiratory failure. Genetic and environmental risk factors have been identified that indicate injury to, and dysfunction of the lung epithelium is central to initiating the pathogenic process. Following injury to the lung epithelium, growth factors, matrikines and extracellular matrix driven signaling together activate a variety of repair pathways that lead to inflammatory cell recruitment, fibroblast proliferation and expansion of the extracellular matrix, culminating in tissue fibrosis. This tissue fibrosis then leads to changes in the biochemical and biomechanical properties of the extracellular matrix, which potentiate profibrotic mechanisms through a "feed-forward cycle." This review provides an overview of the interactions of the pathogenic mechanisms of IPF with a focus on epithelial-mesenchymal crosstalk and the extracellular matrix as a therapeutic target for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Justin C Hewlett
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Veterans Affairs Medical Center, Nashville, TN, United States.
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Veterans Affairs Medical Center, Nashville, TN, United States; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, United States.
| |
Collapse
|