1
|
Maduray K, Zhong J. Emerging roles of ketone bodies in cardiac fibrosis. Am J Physiol Cell Physiol 2024; 327:C1416-C1432. [PMID: 39401423 DOI: 10.1152/ajpcell.00241.2024] [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: 04/14/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 12/11/2024]
Abstract
Cardiac fibrosis, characterized by excessive extracellular matrix (ECM) deposition within the myocardium, poses a significant challenge in cardiovascular health, contributing to various cardiac pathologies. Ketone bodies (KBs), particularly β-hydroxybutyrate (β-OHB), have emerged as subjects of interest due to their potential cardioprotective effects. However, their specific influence on cardiac fibrosis remains underexplored. This literature review comprehensively examines the relationship between KBs and cardiac fibrosis, elucidating potential mechanisms through which KBs modulate fibrotic pathways. A multifaceted interplay exists between KBs and key mediators of cardiac fibrosis. While some studies indicate a profibrotic role for KBs, others highlight their potential to attenuate fibrosis and cardiac remodeling. Mechanistically, KBs may regulate fibrotic pathways through modulation of cellular components such as cardiac fibroblasts, macrophages, and lymphocytes, as well as extracellular matrix proteins. Furthermore, the impact of KBs on cellular processes implicated in fibrosis, including oxidative stress, chemokine and cytokine expression, caspase activation, and inflammasome signaling is explored. While conflicting findings exist regarding the effects of KBs on these processes, emerging evidence suggests a predominantly beneficial role in mitigating inflammation and oxidative stress associated with fibrotic remodeling. Overall, this review underscores the importance of elucidating the complex interplay between KB metabolism and cardiac fibrosis. The insights gained have the potential to inform novel therapeutic strategies for managing cardiac fibrosis and associated cardiovascular disorders, highlighting the need for further research in this area.
Collapse
Affiliation(s)
- Kellina Maduray
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Jingquan Zhong
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Qilu Hospital of Shandong University (Qingdao), Shandong University, Qingdao, Shandong, China
| |
Collapse
|
2
|
Xu W, Chen H, Xiao H. mTORC2: A neglected player in aging regulation. J Cell Physiol 2024; 239:e31363. [PMID: 38982866 DOI: 10.1002/jcp.31363] [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: 03/20/2024] [Revised: 05/21/2024] [Accepted: 06/19/2024] [Indexed: 07/11/2024]
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that plays a pivotal role in various biological processes, through integrating external and internal signals, facilitating gene transcription and protein translation, as well as by regulating mitochondria and autophagy functions. mTOR kinase operates within two distinct protein complexes known as mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), which engage separate downstream signaling pathways impacting diverse cellular processes. Although mTORC1 has been extensively studied as a pro-proliferative factor and a pro-aging hub if activated aberrantly, mTORC2 received less attention, particularly regarding its implication in aging regulation. However, recent studies brought increasing evidence or clues for us, which implies the associations of mTORC2 with aging, as the genetic elimination of unique subunits of mTORC2, such as RICTOR, has been shown to alleviate aging progression in comparison to mTORC1 inhibition. In this review, we first summarized the basic characteristics of mTORC2, including its protein architecture and signaling network. We then focused on reviewing the molecular signaling regulation of mTORC2 in cellular senescence and organismal aging, and proposed the multifaceted regulatory characteristics under senescent and nonsenescent contexts. Next, we outlined the research progress of mTOR inhibitors in the field of antiaging and discussed future prospects and challenges. It is our pleasure if this review article could provide meaningful information for our readers and call forth more investigations working on this topic.
Collapse
Affiliation(s)
- Weitong Xu
- The Lab of Aging Research, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Honghan Chen
- The Lab of Aging Research, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hengyi Xiao
- The Lab of Aging Research, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Wei JY, Ma LX, Liu WT, Dong LH, Hou X, Bao XY, Hou W. Mechanisms and protective measures for radiation-induced brachial plexus nerve injury. Brain Res Bull 2024; 210:110924. [PMID: 38460911 DOI: 10.1016/j.brainresbull.2024.110924] [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: 10/07/2023] [Revised: 02/06/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
Radiation therapy is a common treatment modality for patients with malignant tumors of the head and neck, chest and axilla. However, radiotherapy inevitably causes damage to normal tissues at the irradiated site, among which damage to the brachial plexus nerve(BP) is a serious adverse effect in patients receiving radiation therapy in the scapular or axillary regions, with clinical manifestations including abnormal sensation, neuropathic pain, and dyskinesia, etc. These adverse effects seriously reduce the living quality of patients and pose obstacles to their prognosis. Therefore, it is important to elucidate the mechanism of radiation induced brachial plexus injury (RIBP) which remains unclear. Current studies have shown that the pathways of radiation-induced BP injury can be divided into two categories: direct injury and indirect injury, and the indirect injury is closely related to the inflammatory response, microvascular damage, cytokine production and other factors causing radiation-induced fibrosis. In this review, we summarize the underlying mechanisms of RIBP occurrence and possible effective methods to prevent and treat RIBP.
Collapse
Affiliation(s)
- Jia Ying Wei
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Li Xin Ma
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Wen Tong Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Li Hua Dong
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Xue Hou
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Xue Ying Bao
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Wei Hou
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
4
|
Roccatello D, Lan HY, Sciascia S, Sethi S, Fornoni A, Glassock R. From inflammation to renal fibrosis: A one-way road in autoimmunity? Autoimmun Rev 2024; 23:103466. [PMID: 37848157 DOI: 10.1016/j.autrev.2023.103466] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 10/13/2023] [Indexed: 10/19/2023]
Abstract
Renal fibrosis is now recognized as a main determinant of renal pathology to include chronic kidney disease. Deposition of pathological matrix in the walls of glomerular capillaries, the interstitial space, and around arterioles predicts and contributes to the functional demise of the nephron and its surrounding vasculature. The recent identification of the major cell populations of fibroblast precursors in the kidney interstitium such as pericytes and tissue-resident mesenchymal stem cells, or bone-marrow-derived macrophages, and in the glomerulus such as podocytes, parietal epithelial and mesangial cells, has enabled the study of the fibrogenic process thought the lens of involved immunological pathways. Besides, a growing body of evidence is supporting the role of the lymphatic system in modulating the immunological response potentially leading to inflammation and ultimately renal damage. These notions have moved our understanding of renal fibrosis to be recognized as a clinical entity and new main player in autoimmunity, impacting directly the management of patients.
Collapse
Affiliation(s)
- Dario Roccatello
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley (North-West Italy), San Giovanni Bosco Hub Hospital, ASL Città di Torino and Department of Clinical and Biological Sciences of the University of Turin, Turin, Italy.
| | - Hui-Yao Lan
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Savino Sciascia
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley (North-West Italy), San Giovanni Bosco Hub Hospital, ASL Città di Torino and Department of Clinical and Biological Sciences of the University of Turin, Turin, Italy
| | - Sanjeev Sethi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Alessia Fornoni
- Peggy and Harold Katz Family Drug Discovery Center, Katz Family Division of Nephrology and Hypertension, Department of Medicine, Miller School of Medicine, University of Miami, Miami, USA
| | - Richard Glassock
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
5
|
Mutsaers SE, Krymskaya VP. Pleural Fibrosis: Now That's What mTORC(ing) About. Am J Respir Cell Mol Biol 2024; 70:8-10. [PMID: 37788451 PMCID: PMC10768835 DOI: 10.1165/rcmb.2023-0327ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/03/2023] [Indexed: 10/05/2023] Open
Affiliation(s)
- Steven E Mutsaers
- Institute for Respiratory Health and School of Biomedical Sciences The University of Western Australia Nedlands, Western Australia, Australia
| | - Vera P Krymskaya
- Department of Medicine, Lung Biology Institute Perelman School of Medicine at the University of Pennsylvania Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Keshava S, Owens S, Qin W, Jeffers A, Kyei P, Komatsu S, Kleam J, Ikebe M, Idell S, Tucker TA. The mTORC2/SGK1/NDRG1 Signaling Axis Is Critical for the Mesomesenchymal Transition of Pleural Mesothelial Cells and the Progression of Pleural Fibrosis. Am J Respir Cell Mol Biol 2024; 70:50-62. [PMID: 37607215 PMCID: PMC10768834 DOI: 10.1165/rcmb.2023-0131oc] [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: 04/06/2023] [Accepted: 08/22/2023] [Indexed: 08/24/2023] Open
Abstract
Progressive lung scarring because of persistent pleural organization often results in pleural fibrosis (PF). This process affects patients with complicated parapneumonic pleural effusions, empyema, and other pleural diseases prone to loculation. In PF, pleural mesothelial cells undergo mesomesenchymal transition (MesoMT) to become profibrotic, characterized by increased expression of α-smooth muscle actin and matrix proteins, including collagen-1. In our previous study, we showed that blocking PI3K/Akt signaling inhibits MesoMT induction in human pleural mesothelial cells (HPMCs) (1). However, the downstream signaling pathways leading to MesoMT induction remain obscure. Here, we investigated the role of mTOR complexes (mTORC1/2) in MesoMT induction. Our studies show that activation of the downstream mediator mTORC1/2 complex is, likewise, a critical component of MesoMT. Specific targeting of mTORC1/2 complex using pharmacological inhibitors such as INK128 and AZD8055 significantly inhibited transforming growth factor β (TGF-β)-induced MesoMT markers in HPMCs. We further identified the mTORC2/Rictor complex as the principal contributor to MesoMT progression induced by TGF-β. Knockdown of Rictor, but not Raptor, attenuated TGF-β-induced MesoMT in these cells. In these studies, we further show that concomitant activation of the SGK1/NDRG1 signaling cascade is essential for inducing MesoMT. Targeting SGK1 and NDRG1 with siRNA and small molecular inhibitors attenuated TGF-β-induced MesoMT in HPMCs. Additionally, preclinical studies in our Streptococcus pneumoniae-mediated mouse model of PF showed that inhibition of mTORC1/2 with INK128 significantly attenuated the progression of PF in subacute and chronic injury. In conclusion, our studies demonstrate that mTORC2/Rictor-mediated activation of SGK1/NDRG1 is critical for MesoMT induction and that targeting this pathway could inhibit or even reverse the progression of MesoMT and PF.
Collapse
Affiliation(s)
| | - Shuzi Owens
- Department of Cellular and Molecular Biology, and
| | - Wenyi Qin
- Department of Cellular and Molecular Biology, and
| | | | - Perpetual Kyei
- Biotechnology Graduate Program, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | | | - Joshua Kleam
- Department of Cellular and Molecular Biology, and
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, and
| | - Steven Idell
- Texas Lung Injury Institute
- Department of Cellular and Molecular Biology, and
| | - Torry A. Tucker
- Texas Lung Injury Institute
- Department of Cellular and Molecular Biology, and
| |
Collapse
|
7
|
DiGiovanni GT, Han W, Sherrill TP, Taylor CJ, Nichols DS, Geis NM, Singha UK, Calvi CL, McCall AS, Dixon MM, Liu Y, Jang JH, Gutor SS, Polosukhin VV, Blackwell TS, Kropski JA, Gokey JJ. Epithelial Yap/Taz are required for functional alveolar regeneration following acute lung injury. JCI Insight 2023; 8:e173374. [PMID: 37676731 PMCID: PMC10629815 DOI: 10.1172/jci.insight.173374] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
A hallmark of idiopathic pulmonary fibrosis (IPF) and other interstitial lung diseases is dysregulated repair of the alveolar epithelium. The Hippo pathway effector transcription factors YAP and TAZ are implicated as essential for type 1 and type 2 alveolar epithelial cell (AT1 and AT2) differentiation in the developing lung, yet aberrant activation of YAP/TAZ is a prominent feature of the dysregulated alveolar epithelium in IPF. In these studies, we sought to define the functional role of YAP/TAZ activity during alveolar regeneration. We demonstrated that Yap and Taz were normally activated in AT2 cells shortly after injury, and deletion of Yap/Taz in AT2 cells led to pathologic alveolar remodeling, failure of AT2-to-AT1 cell differentiation, increased collagen deposition, exaggerated neutrophilic inflammation, and increased mortality following injury induced by a single dose of bleomycin. Loss of Yap/Taz activity prior to an LPS injury prevented AT1 cell regeneration, led to intraalveolar collagen deposition, and resulted in persistent innate inflammation. These findings establish that AT2 cell Yap/Taz activity is essential for functional alveolar epithelial repair and prevention of fibrotic remodeling.
Collapse
Affiliation(s)
- Gianluca T. DiGiovanni
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wei Han
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Taylor P. Sherrill
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Chase J. Taylor
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David S. Nichols
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Natalie M. Geis
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ujjal K. Singha
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carla L. Calvi
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - A. Scott McCall
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Molly M. Dixon
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yang Liu
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ji-Hoon Jang
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sergey S. Gutor
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Vasiliy V. Polosukhin
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Jason J. Gokey
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
8
|
Ding M, Shao Y, Sun D, Meng S, Zang Y, Zhou Y, Li J, Lu W, Zhu S. Design, synthesis, and biological evaluation of BRD4 degraders. Bioorg Med Chem 2023; 78:117134. [PMID: 36563515 DOI: 10.1016/j.bmc.2022.117134] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Epigenetic proteins are one of the important targets in the current research fields of cancer therapy. A family of bromodomain-containing (BRD) and extra terminal domain (BET) proteins act as epigenetic readers to regulate the expression of key oncogenes and anti-apoptotic proteins. Recently, although BET degraders based on PROTAC technology have achieved significant antitumor effects, the lack of selectivity for BET protein degradation has not been fully addressed. Herein, a series of small molecule BRD4 PROTACs were designed and synthesized. Most of the degraders were effective in inhibiting MM.1S and MV-4-11 cell lines, especially in MV-4-11. Among them, degrader 8b could induce the degradation of BRD4 and exhibited a time- and concentration-dependent depletion manner and there was a significant depletion of BRD4, laying a foundation for effectively treating leukemia and multiple myeloma. Moreover, 8b could also effectively prevent the activation of MRC5 cells by inducing the degradation of BRD4 protein, which preliminarily proves that the BRD4 degrader based on the PROTAC concept has great potential for the treatment of pulmonary fibrosis. Taken together, these findings laid a foundation for BRD4 degraders as an effective strategy for treating related diseases.
Collapse
Affiliation(s)
- Mengyuan Ding
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Yingying Shao
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Danwen Sun
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Suorina Meng
- University of Chinese Academy of Sciences, NO.19A Yuquan Road, Beijing 100049, PR China; Lingang Laboratory, Shanghai 201203, PR China
| | - Yi Zang
- University of Chinese Academy of Sciences, NO.19A Yuquan Road, Beijing 100049, PR China; Lingang Laboratory, Shanghai 201203, PR China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
| | - Yubo Zhou
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, PR China
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, PR China.
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China.
| | - Shulei Zhu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China.
| |
Collapse
|
9
|
Drake C, Wehr MM, Zobl W, Koschmann J, De Lucca D, Kühne BA, Hansen T, Knebel J, Ritter D, Boei J, Vrieling H, Bitsch A, Escher SE. Substantiate a read-across hypothesis by using transcriptome data-A case study on volatile diketones. FRONTIERS IN TOXICOLOGY 2023; 5:1155645. [PMID: 37206915 PMCID: PMC10188990 DOI: 10.3389/ftox.2023.1155645] [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: 01/31/2023] [Accepted: 04/17/2023] [Indexed: 05/21/2023] Open
Abstract
This case study explores the applicability of transcriptome data to characterize a common mechanism of action within groups of short-chain aliphatic α-, β-, and γ-diketones. Human reference in vivo data indicate that the α-diketone diacetyl induces bronchiolitis obliterans in workers involved in the preparation of microwave popcorn. The other three α-diketones induced inflammatory responses in preclinical in vivo animal studies, whereas beta and gamma diketones in addition caused neuronal effects. We investigated early transcriptional responses in primary human bronchiolar (PBEC) cell cultures after 24 h and 72 h of air-liquid exposure. Differentially expressed genes (DEGs) were assessed based on transcriptome data generated with the EUToxRisk gene panel of Temp-O-Seq®. For each individual substance, genes were identified displaying a consistent differential expression across dose and exposure duration. The log fold change values of the DEG profiles indicate that α- and β-diketones are more active compared to γ-diketones. α-diketones in particular showed a highly concordant expression pattern, which may serve as a first indication of the shared mode of action. In order to gain a better mechanistic understanding, the resultant DEGs were submitted to a pathway analysis using ConsensusPathDB. The four α-diketones showed very similar results with regard to the number of activated and shared pathways. Overall, the number of signaling pathways decreased from α-to β-to γ-diketones. Additionally, we reconstructed networks of genes that interact with one another and are associated with different adverse outcomes such as fibrosis, inflammation or apoptosis using the TRANSPATH-database. Transcription factor enrichment and upstream analyses with the geneXplain platform revealed highly interacting gene products (called master regulators, MRs) per case study compound. The mapping of the resultant MRs on the reconstructed networks, visualized similar gene regulation with regard to fibrosis, inflammation and apoptosis. This analysis showed that transcriptome data can strengthen the similarity assessment of compounds, which is of particular importance, e.g., in read-across approaches. It is one important step towards grouping of compounds based on biological profiles.
Collapse
Affiliation(s)
- Christina Drake
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
- *Correspondence: Christina Drake,
| | - Matthias M. Wehr
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Walter Zobl
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | | | | | - Britta A. Kühne
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Tanja Hansen
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Jan Knebel
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Detlef Ritter
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Jan Boei
- Leiden University Medical Center, Leiden, Netherlands
| | | | - Annette Bitsch
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Sylvia E. Escher
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| |
Collapse
|
10
|
Huang J, Chen L, Wu J, Ai D, Zhang JQ, Chen TG, Wang L. Targeting the PI3K/AKT/mTOR Signaling Pathway in the Treatment of Human Diseases: Current Status, Trends, and Solutions. J Med Chem 2022; 65:16033-16061. [PMID: 36503229 DOI: 10.1021/acs.jmedchem.2c01070] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway is one of the most important intracellular pathways involved in cell proliferation, growth, differentiation, and survival. Therefore, this route is a prospective biological target for treating various human diseases, such as tumors, neurodegenerative diseases, pulmonary fibrosis, and diabetes. An increasing number of clinical studies emphasize the necessity of developing novel molecules targeting the PI3K/AKT/mTOR pathway. This review focuses on recent advances in ATP-competitive inhibitors, allosteric inhibitors, covalent inhibitors, and proteolysis-targeting chimeras against the PI3K/AKT/mTOR pathway, and highlights possible solutions for overcoming the toxicities and acquired drug resistance of currently available drugs. We also provide recommendations for the future design and development of promising drugs targeting this pathway.
Collapse
Affiliation(s)
- Jindi Huang
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Liye Chen
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiangxia Wu
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Daiqiao Ai
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Ji-Quan Zhang
- College of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Tie-Gen Chen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Room 109, Building C, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Ling Wang
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
11
|
Caracena T, Blomberg R, Hewawasam RS, Fry ZE, Riches DWH, Magin CM. Alveolar epithelial cells and microenvironmental stiffness synergistically drive fibroblast activation in three-dimensional hydrogel lung models. Biomater Sci 2022; 10:7133-7148. [PMID: 36366982 PMCID: PMC9729409 DOI: 10.1039/d2bm00827k] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease that progressively and irreversibly alters the lung parenchyma, eventually leading to respiratory failure. The study of this disease has been historically challenging due to the myriad of complex processes that contribute to fibrogenesis and the inherent difficulty in accurately recreating the human pulmonary environment in vitro. Here, we describe a poly(ethylene glycol) PEG hydrogel-based three-dimensional model for the co-culture of primary murine pulmonary fibroblasts and alveolar epithelial cells that reproduces the micro-architecture, cell placement, and mechanical properties of healthy and fibrotic lung tissue. Co-cultured cells retained normal levels of viability up to at least three weeks and displayed differentiation patterns observed in vivo during IPF progression. Interrogation of protein and gene expression within this model showed that myofibroblast activation required both extracellular mechanical cues and the presence of alveolar epithelial cells. Differences in gene expression indicated that cellular co-culture induced TGF-β signaling and proliferative gene expression, while microenvironmental stiffness upregulated the expression of genes related to cell-ECM interactions. This biomaterial-based cell culture system serves as a significant step forward in the accurate recapitulation of human lung tissue in vitro and highlights the need to incorporate multiple factors that work together synergistically in vivo into models of lung biology of health and disease.
Collapse
Affiliation(s)
- Thomas Caracena
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, USA.
| | - Rachel Blomberg
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, USA.
| | - Rukshika S Hewawasam
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, USA.
| | - Zoe E Fry
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, USA.
| | - David W H Riches
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, USA
- Department of Research, Veterans Affairs Eastern Colorado Health Care System, USA
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, USA
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, USA.
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, USA
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, USA
| |
Collapse
|
12
|
Liu H, Pang Q, Cao F, Liu Z, Wei W, Li Z, Long Q, Jiao Y. Number 2 Feibi Recipe Ameliorates Pulmonary Fibrosis by Inducing Autophagy Through the GSK-3β/mTOR Pathway. Front Pharmacol 2022; 13:921209. [PMID: 35903328 PMCID: PMC9315309 DOI: 10.3389/fphar.2022.921209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Number 2 Feibi Recipe (N2FBR) is a traditional Chinese medicine formula for treating idiopathic pulmonary fibrosis. N2FBR inhibits H2O2-mediated oxidative stress damage in alveolar epithelial cells by increasing autophagy, as we previously demonstrated. However, it is unknown if similar mechanisms occur in vivo. We established a pulmonary fibrosis model by instilling bleomycin (BLM) from the airway to examine the effects of N2FBR on pulmonary fibrosis and investigate its probable mechanism in this work. We discovered that N2FBR treatment effectively alleviated interstitial fibrosis as well as collagen deposition, primarily in upregulating SOD, GSH-Px, T-AOC and downregulating MDA content. N2FBR also increased the expression of LC3B, Beclin-1, LAMP1, TFEB and downregulated the expression of p62, legumain. N2FBR treatment boosted the production of autophagosomes, according to the results of the TEM observation. Furthermore, we explored that N2FBR exerted its anti-oxidative stress and pro-autophagy effects via GSK-3β/mTOR signalling pathway. Therefore, these results provide further evidence for the protective effect of N2FBR in pulmonary fibrosis. Our findings could have ramifications for the development of antifibrosis therapies.
Collapse
Affiliation(s)
- Haoge Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qinglu Pang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Fang Cao
- Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhaoheng Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Wan Wei
- Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhipeng Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Long
- Department of Respiratory and Critical Care Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
- *Correspondence: Qi Long, ; Yang Jiao,
| | - Yang Jiao
- Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Qi Long, ; Yang Jiao,
| |
Collapse
|
13
|
Gokey JJ, Patel SD, Kropski JA. The Role of Hippo/YAP Signaling in Alveolar Repair and Pulmonary Fibrosis. Front Med (Lausanne) 2021; 8:752316. [PMID: 34671628 PMCID: PMC8520933 DOI: 10.3389/fmed.2021.752316] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/09/2021] [Indexed: 01/30/2023] Open
Abstract
Pulmonary fibrosis is characterized by loss of normal alveoli, accumulation of pathologic activated fibroblasts, and exuberant extracellular matrix deposition that over time can lead to progressive loss of respiratory function and death. This loss of respiratory function is associated with the loss of alveolar type 1 cells (AT1) that play a crucial role in gas exchange and the depletion of the alveolar type 2 cells (AT2) that act as progenitor cells to regenerate the AT1 and AT2 cell populations during repair. Understanding the mechanisms that regulate normal alveolar repair and those associated with pathologic repair is essential to identify potential therapeutic targets to treat or delay progression of fibrotic diseases. The Hippo/YAP developmental signaling pathway has been implicated as a regulator of normal alveolar development and repair. In idiopathic pulmonary fibrosis, aberrant activation of YAP/TAZ has been demonstrated in both the alveolar epithelium and activated fibroblasts associated with increased fibrotic remodeling, and there is emerging interest in this pathway as a target for antifibrotic therapies. In this review, we summarize current evidence as to the role of the Hippo-YAP/TAZ pathway in alveolar development, homeostasis, and repair, and highlight key questions that must be resolved to determine effective strategies to modulate YAP/TAZ signaling to prevent progressive pulmonary fibrosis and enhance adaptive alveolar repair.
Collapse
Affiliation(s)
- Jason J Gokey
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Saawan D Patel
- 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 Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States.,Department of Veterans Affairs Medical Center, Nashville, TN, United States
| |
Collapse
|
14
|
Liu H, Liu C, Wang M, Sun D, Zhu P, Zhang P, Tan X, Shi G. Tanshinone IIA affects the malignant growth of Cholangiocarcinoma cells by inhibiting the PI3K-Akt-mTOR pathway. Sci Rep 2021; 11:19268. [PMID: 34588580 PMCID: PMC8481305 DOI: 10.1038/s41598-021-98948-z] [Citation(s) in RCA: 4] [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: 05/19/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
In the present study, we aimed to find the target of Tanshinone IIA (Tan-IIA) in Cholangiocarcinoma by network pharmacology-based prediction and investigate the possible mechanism through experimental verification. In this study, we combined Tan-IIA-specific and Cholangiocarcinoma-specific targets with protein-protein interactions (PPI) to construct a Tan-IIA targets-Cholangiocarcinoma network, and network pharmacology approach was applied to identify potential targets and mechanisms of Tan-IIA in the treatment of Cholangiocarcinoma. The anti-cancer effects of Tan-IIA were investigated by using subcutaneous tumorigenic model in nude mice and in the human Cholangiocarcinoma cell lines in vitro. Our results showed that Tan-IIA treatment considerably suppressed the proliferation and migration of Cholangiocarcinoma cells while inducing apoptosis of Cholangiocarcinoma cells. Western blot results demonstrated that the expression of PI3K, p-Akt, p-mTOR, and mTOR were inhibited by Tan-IIA. Meanwhile, After treatment with Tan-IIA, the level of Bcl2 was downregulated and cleaved caspase-3 expression increased. Further studies revealed that the anticancer effects of Tan-IIA were severely mitigated by pretreatment with a PI3K agonist. Our research provides a new anticancer strategy and strengthens support for the use of Tan-IIA as an anticancer drug for the treatment of CCA.
Collapse
Affiliation(s)
- Huayuan Liu
- Department of Medicine, Qingdao University, Qingdao, China
| | - Caiyun Liu
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Mengya Wang
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Dongxu Sun
- Graduate School of Dalian Medical University, Dalian, China
| | - Pengcheng Zhu
- Graduate School of Dalian Medical University, Dalian, China
| | - Ping Zhang
- Department of Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Xueying Tan
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Guangjun Shi
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
15
|
Selvarajah B, Azuelos I, Anastasiou D, Chambers RC. Fibrometabolism-An emerging therapeutic frontier in pulmonary fibrosis. Sci Signal 2021; 14:14/697/eaay1027. [PMID: 34429381 DOI: 10.1126/scisignal.aay1027] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Fibrosis is the final pathological outcome and major cause of morbidity and mortality in many common and chronic inflammatory, immune-mediated, and metabolic diseases. Despite the growing incidence of fibrotic diseases and extensive research efforts, there remains a lack of effective therapies that improve survival. The application of omics technologies has revolutionized our approach to identifying previously unknown therapeutic targets and potential disease biomarkers. The application of metabolomics, in particular, has improved our understanding of disease pathomechanisms and garnered a wave of scientific interest in the role of metabolism in the biology of myofibroblasts, the key effector cells of the fibrogenic response. Emerging evidence suggests that alterations in metabolism not only are a feature of but also may play an influential role in the pathogenesis of fibrosis, most notably in idiopathic pulmonary fibrosis (IPF), the most rapidly progressive and fatal of all fibrotic conditions. This review will detail the role of key metabolic pathways, their alterations in myofibroblasts, and the potential this new knowledge offers for the development of antifibrotic therapeutic strategies.
Collapse
Affiliation(s)
- Brintha Selvarajah
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Ilan Azuelos
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London WC1E 6JF, UK
| | | | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London WC1E 6JF, UK.
| |
Collapse
|
16
|
Liu T, Song Y, Hu A. Neuroprotective mechanisms of mangiferin in neurodegenerative diseases. Drug Dev Res 2021; 82:494-502. [PMID: 33458836 DOI: 10.1002/ddr.21783] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 11/10/2022]
Abstract
The central nervous system (CNS) regulates and coordinates an extensive array of complex processes requiring harmonious regulation of specific genes. CNS disorders represent a large burden on society and cause enormous disability and economic losses. Traditional Chinese medicine (TCM) has been used for many years in the treatment of neurological illnesses, such as Alzheimer's disease, Parkinson's disease, stroke, and depression, as the combination of TCM and Western medicine has superior therapeutic efficacy and minimal toxic side effects. Mangiferin (MGF) is an active compound of the traditional Chinese herb rhizome anemarrhenae, which has antioxidant, anti-inflammation, anti-lipid peroxidation, immunomodulatory, and anti-apoptotic functions in the CNS. MGF has been demonstrated to have therapeutic effects in CNS diseases through a multitude of mechanisms. This review outlines the latest research on the neuroprotective ability of MGF and the diverse molecular mechanisms involved.
Collapse
Affiliation(s)
- Tingjun Liu
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, PR China
| | - Yuanjian Song
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China.,Department of Genetics, Xuzhou Engineering Research Center of Medical Genetics and Transformation, Xuzhou Medical University, Xuzhou, China
| | - Ankang Hu
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, PR China
| |
Collapse
|
17
|
O'Leary EM, Tian Y, Nigdelioglu R, Witt LJ, Cetin-Atalay R, Meliton AY, Woods PS, Kimmig LM, Sun KA, Gökalp GA, Mutlu GM, Hamanaka RB. TGF-β Promotes Metabolic Reprogramming in Lung Fibroblasts via mTORC1-dependent ATF4 Activation. Am J Respir Cell Mol Biol 2020; 63:601-612. [PMID: 32668192 DOI: 10.1165/rcmb.2020-0143oc] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a fatal interstitial lung disease characterized by the TGF-β (transforming growth factor-β)-dependent differentiation of lung fibroblasts into myofibroblasts, which leads to excessive deposition of collagen proteins and progressive scarring. We have previously shown that synthesis of collagen by myofibroblasts requires de novo synthesis of glycine, the most abundant amino acid found in collagen protein. TGF-β upregulates the expression of the enzymes of the de novo serine-glycine synthesis pathway in lung fibroblasts; however, the transcriptional and signaling regulators of this pathway remain incompletely understood. Here, we demonstrate that TGF-β promotes accumulation of ATF4 (activating transcription factor 4), which is required for increased expression of the serine-glycine synthesis pathway enzymes in response to TGF-β. We found that induction of the integrated stress response (ISR) contributes to TGF-β-induced ATF4 activity; however, the primary driver of ATF4 downstream of TGF-β is activation of mTORC1 (mTOR Complex 1). TGF-β activates the PI3K-Akt-mTOR pathway, and inhibition of PI3K prevents activation of downstream signaling and induction of ATF4. Using a panel of mTOR inhibitors, we found that ATF4 activation is dependent on mTORC1, independent of mTORC2. Rapamycin, which incompletely and allosterically inhibits mTORC1, had no effect on TGF-β-mediated induction of ATF4; however, Rapalink-1, which specifically targets the kinase domain of mTORC1, completely inhibited ATF4 induction and metabolic reprogramming downstream of TGF-β. Our results provide insight into the mechanisms of metabolic reprogramming in myofibroblasts and clarify contradictory published findings on the role of mTOR inhibition in myofibroblast differentiation.
Collapse
Affiliation(s)
- Erin M O'Leary
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Yufeng Tian
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Recep Nigdelioglu
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois; and
| | - Leah J Witt
- Division of Geriatrics and Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, The University of California San Francisco, San Francisco, California
| | - Rengul Cetin-Atalay
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Angelo Y Meliton
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Parker S Woods
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Lucas M Kimmig
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Kaitlyn A Sun
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Gizem A Gökalp
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Gökhan M Mutlu
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Robert B Hamanaka
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| |
Collapse
|
18
|
Rackow AR, Nagel DJ, McCarthy C, Judge J, Lacy S, Freeberg MAT, Thatcher TH, Kottmann RM, Sime PJ. The self-fulfilling prophecy of pulmonary fibrosis: a selective inspection of pathological signalling loops. Eur Respir J 2020; 56:13993003.00075-2020. [PMID: 32943406 PMCID: PMC7931159 DOI: 10.1183/13993003.00075-2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/01/2020] [Indexed: 12/28/2022]
Abstract
Pulmonary fibrosis is a devastating, progressive disease and carries a prognosis worse than most cancers. Despite ongoing research, the mechanisms that underlie disease pathogenesis remain only partially understood. However, the self-perpetuating nature of pulmonary fibrosis has led several researchers to propose the existence of pathological signalling loops. According to this hypothesis, the normal wound-healing process becomes corrupted and results in the progressive accumulation of scar tissue in the lung. In addition, several negative regulators of pulmonary fibrosis are downregulated and, therefore, are no longer capable of inhibiting these feed-forward loops. The combination of pathological signalling loops and loss of a checks and balances system ultimately culminates in a process of unregulated scar formation. This review details specific signalling pathways demonstrated to play a role in the pathogenesis of pulmonary fibrosis. The evidence of detrimental signalling loops is elucidated with regard to epithelial cell injury, cellular senescence and the activation of developmental and ageing pathways. We demonstrate where these loops intersect each other, as well as common mediators that may drive these responses and how the loss of pro-resolving mediators may contribute to the propagation of disease. By focusing on the overlapping signalling mediators among the many pro-fibrotic pathways, it is our hope that the pulmonary fibrosis community will be better equipped to design future trials that incorporate the redundant nature of these pathways as we move towards finding a cure for this unrelenting disease.
Collapse
Affiliation(s)
- Ashley R Rackow
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Authors contributed equally to this work
| | - David J Nagel
- Division of Pulmonary Diseases and Critical Care, University of Rochester Medical Center, Rochester, NY, USA.,Authors contributed equally to this work
| | | | | | - Shannon Lacy
- US Army of Veterinary Corps, Fort Campbell, KY, USA
| | | | - Thomas H Thatcher
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - R Matthew Kottmann
- Division of Pulmonary Diseases and Critical Care, University of Rochester Medical Center, Rochester, NY, USA
| | - Patricia J Sime
- Division of Pulmonary Diseases and Critical Care, University of Rochester Medical Center, Rochester, NY, USA.,Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
19
|
Clynick B, Jo HE, Corte TJ, Glaspole IN, Grainge C, Hopkins PMA, Reynolds PN, Chapman S, Walters EH, Zappala C, Keir GJ, Cooper WA, Mahar AM, Ellis S, Goh NS, Baltic S, Ryan M, Tan DBA, Moodley YP. Circulating RNA differences between patients with stable and progressive idiopathic pulmonary fibrosis. Eur Respir J 2020; 56:13993003.02058-2019. [PMID: 32381494 DOI: 10.1183/13993003.02058-2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/08/2020] [Indexed: 11/05/2022]
Affiliation(s)
- Britt Clynick
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia
| | - Helen E Jo
- University of Sydney, Camperdown, Australia.,Royal Prince Alfred Hospital, Camperdown, Australia
| | - Tamera J Corte
- University of Sydney, Camperdown, Australia.,Royal Prince Alfred Hospital, Camperdown, Australia
| | - Ian N Glaspole
- Monash University, Clayton, Australia.,Alfred Hospital, Melbourne, Australia
| | - Christopher Grainge
- University of Newcastle, Callaghan, Australia.,John Hunter Hospital, New Lambton Heights, Australia
| | - Peter M A Hopkins
- University of Queensland, St Lucia, Australia.,Prince Charles Hospital, Chermside, Australia
| | - Paul N Reynolds
- University of Adelaide, Adelaide, Australia.,Royal Adelaide Hospital, Adelaide, Australia
| | | | - E Haydn Walters
- Alfred Hospital, Melbourne, Australia.,University of Tasmania, Hobart, Australia.,University of Melbourne, Parkville, Australia.,Royal Hobart Hospital, Hobart, Australia
| | | | | | - Wendy A Cooper
- University of Sydney, Camperdown, Australia.,Royal Prince Alfred Hospital, Camperdown, Australia.,Western Sydney University, Sydney, Australia
| | - Annabelle M Mahar
- University of Sydney, Camperdown, Australia.,Royal Prince Alfred Hospital, Camperdown, Australia
| | | | - Nicole S Goh
- Austin Hospital, Heidelberg, Australia.,Institute of Breathing and Sleep, Heidelberg, Australia
| | - Svetlana Baltic
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia
| | - Marisa Ryan
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia
| | - Dino B A Tan
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia
| | - Yuben P Moodley
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia.,Fiona Stanley Hospital, Murdoch, Australia
| |
Collapse
|
20
|
Ren R, Ruan Z, Ding H, Du J, Yu W. Phosphoproteome profiling provides insight into the mechanisms of ventilator-induced lung injury. Exp Ther Med 2020; 19:3627-3633. [PMID: 32346427 PMCID: PMC7185165 DOI: 10.3892/etm.2020.8634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
The incidence of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) is a common health problem in the clinic and is projected to increase in prevalence in the future. Mechanical ventilation is commonly used to provide respiratory support and has become indispensable in emergency and critical medicine. However, ventilator use can result in lung tissue damage, collectively termed ventilator-induced lung injury (VILI). In the present study, phosphoprotein profiling of blood and tissue samples from ventilated and non-ventilated mice was performed and key changes in protein levels and cell signaling during VILI were identified. Activation of the PI3K/AKT and mitogen activated protein kinase signaling pathways, in addition to changes in expression of cancer, inflammatory and cell-death related proteins were detected in response to mechanical ventilation. Focal adhesion-related protein levels and signaling pathways were also significantly altered in an injury model compared with control. VILI can affect patient mortality in ALI and ARDS cases, and no targeted treatment options currently exist. Identifying biomarkers and understanding the signaling pathways associated with VILI is critical for the development of future therapies.
Collapse
Affiliation(s)
- Rongrong Ren
- Department of Anesthesiology, The Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, P.R. China.,Department of Anesthesiology and Surgical Intensive Care Unit, Xin Hua Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Zhengshang Ruan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xin Hua Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Haoshu Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xin Hua Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Junming Du
- Department of Anesthesiology and Surgical Intensive Care Unit, Xin Hua Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Weifeng Yu
- Department of Anesthesiology, The Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| |
Collapse
|
21
|
Farhood B, Khodamoradi E, Hoseini-Ghahfarokhi M, Motevaseli E, Mirtavoos-Mahyari H, Eleojo Musa A, Najafi M. TGF-β in radiotherapy: Mechanisms of tumor resistance and normal tissues injury. Pharmacol Res 2020; 155:104745. [PMID: 32145401 DOI: 10.1016/j.phrs.2020.104745] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/25/2020] [Accepted: 03/03/2020] [Indexed: 12/20/2022]
Abstract
Emerging evidences show that changes in tumor stroma can adapt cancer cells to radiotherapy, thereby leading to a reduction in tumor response to treatment. On the other hand, radiotherapy is associated with severe reactions in normal tissues which limit the amount radiation dose received by tumor. These challenges open a window in radiobiology and radiation oncology to explore mechanisms for improving tumor response and also alleviate side effects of radiotherapy. Transforming growth factor beta (TGF-β) is a well-known and multitasking cytokine that regulates a wide range of reactions and interactions within tumor and normal tissues. Within tumor microenvironment (TME), TGF-β is the most potent suppressor of immune system activity against cancer cells. This effect is mediated through stimulation of CD4+ which differentiates to T regulatory cells (Tregs), infiltration of fibroblasts and differentiation into cancer associated fibroblasts (CAFs), and also polarization of macrophages to M2 cells. These changes lead to suppression of cytotoxic CD8 + T lymphocytes (CTLs) and natural killer (NK) cells to kill cancer cells. TGF-β also plays a key role in the angiogenesis, invasion and DNA damage responses (DDR) in cancer cells. In normal tissues, TGF-β triggers the expression of a wide range of pro-oxidant and pro-fibrosis genes, leading to fibrosis, genomic instability and some other side effects. These properties of TGF-β make it a potential target to preserve normal tissues and sensitize tumor via its inhibition. In the current review, we aim to explain the mechanisms of upregulation of TGF-β and its consequences in both tumor and normal tissues.
Collapse
Affiliation(s)
- Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojtaba Hoseini-Ghahfarokhi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanifeh Mirtavoos-Mahyari
- Lung Transplantation Research Center (LTRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
22
|
P27 Promotes TGF- β-Mediated Pulmonary Fibrosis via Interacting with MTORC2. Can Respir J 2019; 2019:7157861. [PMID: 31641391 PMCID: PMC6770332 DOI: 10.1155/2019/7157861] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022] Open
Abstract
Pulmonary fibrosis (PF), a progressive and life-threatening pulmonary disease, is the main pathological basis of interstitial lung disease (ILD) which includes the idiopathic pulmonary fibrosis (IPF). No effective therapeutic strategy for pulmonary fibrosis has been established. TGF-β signaling has emerged as the vital regulator of PF; however, the detailed molecular mechanisms of TGF-β in PF were uncertain. In the present study, we proved that inhibition of MTORC2 suppresses the expression of P27 in MRC5 and HLF cells. And in bleomycin-induced PF model, the expression of α-SMA and P27 was upregulated. Moreover, TGF-β application increased the level of α-SMA, vimentin, and P27 in MRC5 and HLF cells. Furthermore, P27 overexpression advanced the cell cycle process and promoted the proliferation of MRC5 and HLF cells. Finally, the rescue experiment showed that MTORC2 knockdown reversed P27 overexpression-induced cell cycle acceleration and proliferation. Thus, our results suggest that P27 is involved in TGF-β-mediated PF, which was regulated by MTORC2, providing a novel insight into the development of PF.
Collapse
|
23
|
Ko J, Mills T, Huang J, Chen NY, Mertens TCJ, Collum SD, Lee G, Xiang Y, Han L, Zhou Y, Lee CG, Elias JA, Jyothula SSK, Rajagopal K, Karmouty-Quintana H, Blackburn MR. Transforming growth factor β1 alters the 3'-UTR of mRNA to promote lung fibrosis. J Biol Chem 2019; 294:15781-15794. [PMID: 31488543 DOI: 10.1074/jbc.ra119.009148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/27/2019] [Indexed: 12/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic disease characterized by the pathological remodeling of air sacs as a result of excessive accumulation of extracellular matrix (ECM) proteins, but the mechanism governing the robust protein expression is poorly understood. Our recent findings demonstrate that alternative polyadenylation (APA) caused by NUDT21 reduction is important for the increased expression of fibrotic mediators and ECM proteins in lung fibroblasts by shortening the 3'-untranslated regions (3'-UTRs) of mRNAs and stabilizing their transcripts, therefore activating pathological signaling pathways. Despite the importance of NUDT21 reduction in the regulation of fibrosis, the underlying mechanisms for the depletion are unknown. We demonstrate here that NUDT21 is depleted by TGFβ1. We found that miR203, which is increased in IPF, was induced by TGFβ1 to target the NUDT21 3'-UTR, thus depleting NUDT21 in human and mouse lung fibroblasts. TGFβ1-mediated NUDT21 reduction was attenuated by the miR203 inhibitor antagomiR203 in fibroblasts. TGFβ1 transgenic mice revealed that TGFβ1 down-regulates NUDT21 in fibroblasts in vivo Furthermore, TGFβ1 promoted differential APA of fibrotic genes, including FGF14, RICTOR, TMOD2, and UCP5, in association with increased protein expression. This unique differential APA signature was also observed in IPF fibroblasts. Altogether, our results identified TGFβ1 as an APA regulator through NUDT21 depletion amplifying pulmonary fibrosis.
Collapse
Affiliation(s)
- Junsuk Ko
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030.,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Jingjing Huang
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210003 Jiangsu, China
| | - Ning-Yuan Chen
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Tinne C J Mertens
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Scott D Collum
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030.,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Garam Lee
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Yu Xiang
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Leng Han
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030.,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Soma S K Jyothula
- Department of Internal Medicine, McGovern Medical School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Keshava Rajagopal
- Department of Internal Medicine, McGovern Medical School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030.,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Michael R Blackburn
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030 .,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| |
Collapse
|
24
|
Selvarajah B, Azuelos I, Platé M, Guillotin D, Forty EJ, Contento G, Woodcock HV, Redding M, Taylor A, Brunori G, Durrenberger PF, Ronzoni R, Blanchard AD, Mercer PF, Anastasiou D, Chambers RC. mTORC1 amplifies the ATF4-dependent de novo serine-glycine pathway to supply glycine during TGF-β 1-induced collagen biosynthesis. Sci Signal 2019; 12:eaav3048. [PMID: 31113850 PMCID: PMC6584619 DOI: 10.1126/scisignal.aav3048] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The differentiation of fibroblasts into a transient population of highly activated, extracellular matrix (ECM)-producing myofibroblasts at sites of tissue injury is critical for normal tissue repair. Excessive myofibroblast accumulation and persistence, often as a result of a failure to undergo apoptosis when tissue repair is complete, lead to pathological fibrosis and are also features of the stromal response in cancer. Myofibroblast differentiation is accompanied by changes in cellular metabolism, including increased glycolysis, to meet the biosynthetic demands of enhanced ECM production. Here, we showed that transforming growth factor-β1 (TGF-β1), the key pro-fibrotic cytokine implicated in multiple fibrotic conditions, increased the production of activating transcription factor 4 (ATF4), the transcriptional master regulator of amino acid metabolism, to supply glucose-derived glycine to meet the amino acid requirements associated with enhanced collagen production in response to myofibroblast differentiation. We further delineated the signaling pathways involved and showed that TGF-β1-induced ATF4 production depended on cooperation between canonical TGF-β1 signaling through Smad3 and activation of mechanistic target of rapamycin complex 1 (mTORC1) and its downstream target eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1). ATF4, in turn, promoted the transcription of genes encoding enzymes of the de novo serine-glycine biosynthetic pathway and glucose transporter 1 (GLUT1). Our findings suggest that targeting the TGF-β1-mTORC1-ATF4 axis may represent a novel therapeutic strategy for interfering with myofibroblast function in fibrosis and potentially in other conditions, including cancer.
Collapse
Affiliation(s)
- Brintha Selvarajah
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Ilan Azuelos
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Manuela Platé
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Delphine Guillotin
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Ellen J Forty
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Greg Contento
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Hannah V Woodcock
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Matthew Redding
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Adam Taylor
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK
| | - Gino Brunori
- GlaxoSmithKline, David Jack Centre for R&D, Park Road, Ware, Hertfordshire, SG12 0DP, UK
| | - Pascal F Durrenberger
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Riccardo Ronzoni
- Centre for Respiratory Biology, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | - Andy D Blanchard
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK
| | - Paul F Mercer
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK
| | | | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London WC1E 6JF, UK.
| |
Collapse
|
25
|
Carvedilol attenuates experimentally induced silicosis in rats via modulation of P-AKT/mTOR/TGFβ1 signaling. Int Immunopharmacol 2019; 70:47-55. [DOI: 10.1016/j.intimp.2019.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/06/2019] [Accepted: 02/06/2019] [Indexed: 12/27/2022]
|
26
|
Summer R, Shaghaghi H, Schriner D, Roque W, Sales D, Cuevas-Mora K, Desai V, Bhushan A, Ramirez MI, Romero F. Activation of the mTORC1/PGC-1 axis promotes mitochondrial biogenesis and induces cellular senescence in the lung epithelium. Am J Physiol Lung Cell Mol Physiol 2019; 316:L1049-L1060. [PMID: 30892080 DOI: 10.1152/ajplung.00244.2018] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cellular senescence is a biological process by which cells lose their capacity to proliferate yet remain metabolically active. Although originally considered a protective mechanism to limit the formation of cancer, it is now appreciated that cellular senescence also contributes to the development of disease, including common respiratory ailments such as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. While many factors have been linked to the development of cellular senescence, mitochondrial dysfunction has emerged as an important causative factor. In this study, we uncovered that the mitochondrial biogenesis pathway driven by the mammalian target of rapamycin/peroxisome proliferator-activated receptor-γ complex 1α/β (mTOR/PGC-1α/β) axis is markedly upregulated in senescent lung epithelial cells. Using two different models, we show that activation of this pathway is associated with other features characteristic of enhanced mitochondrial biogenesis, including elevated number of mitochondrion per cell, increased oxidative phosphorylation, and augmented mitochondrial reactive oxygen species (ROS) production. Furthermore, we found that pharmacological inhibition of the mTORC1 complex with rapamycin not only restored mitochondrial homeostasis but also reduced cellular senescence to bleomycin in lung epithelial cells. Likewise, mitochondrial-specific antioxidant therapy also effectively inhibited mTORC1 activation in these cells while concomitantly reducing mitochondrial biogenesis and cellular senescence. In summary, this study provides a mechanistic link between mitochondrial biogenesis and cellular senescence in lung epithelium and suggests that strategies aimed at blocking the mTORC1/PGC-1α/β axis or reducing ROS-induced molecular damage could be effective in the treatment of senescence-associated lung diseases.
Collapse
Affiliation(s)
- Ross Summer
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Hoora Shaghaghi
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - DeLeila Schriner
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Willy Roque
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Dominic Sales
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Karina Cuevas-Mora
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Vilas Desai
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Alok Bhushan
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Maria I Ramirez
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Freddy Romero
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Woodcock HV, Eley JD, Guillotin D, Platé M, Nanthakumar CB, Martufi M, Peace S, Joberty G, Poeckel D, Good RB, Taylor AR, Zinn N, Redding M, Forty EJ, Hynds RE, Swanton C, Karsdal M, Maher TM, Fisher A, Bergamini G, Marshall RP, Blanchard AD, Mercer PF, Chambers RC. The mTORC1/4E-BP1 axis represents a critical signaling node during fibrogenesis. Nat Commun 2019; 10:6. [PMID: 30602778 PMCID: PMC6315032 DOI: 10.1038/s41467-018-07858-8] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 11/29/2018] [Indexed: 01/06/2023] Open
Abstract
Myofibroblasts are the key effector cells responsible for excessive extracellular matrix deposition in multiple fibrotic conditions, including idiopathic pulmonary fibrosis (IPF). The PI3K/Akt/mTOR axis has been implicated in fibrosis, with pan-PI3K/mTOR inhibition currently under clinical evaluation in IPF. Here we demonstrate that rapamycin-insensitive mTORC1 signaling via 4E-BP1 is a critical pathway for TGF-β1 stimulated collagen synthesis in human lung fibroblasts, whereas canonical PI3K/Akt signaling is not required. The importance of mTORC1 signaling was confirmed by CRISPR-Cas9 gene editing in normal and IPF fibroblasts, as well as in lung cancer-associated fibroblasts, dermal fibroblasts and hepatic stellate cells. The inhibitory effect of ATP-competitive mTOR inhibition extended to other matrisome proteins implicated in the development of fibrosis and human disease relevance was demonstrated in live precision-cut IPF lung slices. Our data demonstrate that the mTORC1/4E-BP1 axis represents a critical signaling node during fibrogenesis with potential implications for the development of novel anti-fibrotic strategies.
Collapse
Affiliation(s)
- Hannah V Woodcock
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London, WC1E 6JF, UK
| | - Jessica D Eley
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London, WC1E 6JF, UK
| | - Delphine Guillotin
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London, WC1E 6JF, UK
| | - Manuela Platé
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London, WC1E 6JF, UK
| | - Carmel B Nanthakumar
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Matteo Martufi
- Target Sciences, Medicines Research Centre, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Simon Peace
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Gerard Joberty
- Cellzome, a GSK Company, Meyershofstrasse 1, 69117, Heidelberg, Germany
| | - Daniel Poeckel
- Cellzome, a GSK Company, Meyershofstrasse 1, 69117, Heidelberg, Germany
| | - Robert B Good
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Adam R Taylor
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Nico Zinn
- Cellzome, a GSK Company, Meyershofstrasse 1, 69117, Heidelberg, Germany
| | - Matthew Redding
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London, WC1E 6JF, UK
| | - Ellen J Forty
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London, WC1E 6JF, UK
| | - Robert E Hynds
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, WC1E 6DD, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Charles Swanton
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, WC1E 6DD, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | | | - Toby M Maher
- Fibrosis Research Group, Inflammation, Repair & Development Section, NHLI, Imperial College, London, SW3 6LY, UK
| | - Andrew Fisher
- Newcastle Fibrosis Research Group, Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | | | - Richard P Marshall
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Andy D Blanchard
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Paul F Mercer
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London, WC1E 6JF, UK
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Rayne Building, University College London, London, WC1E 6JF, UK.
| |
Collapse
|
28
|
Xi JS, Wang YF, Long XX, Ma Y. Mangiferin Potentiates Neuroprotection by Isoflurane in Neonatal Hypoxic Brain Injury by Reducing Oxidative Stress and Activation of Phosphatidylinositol-3-Kinase/Akt/Mammalian Target of Rapamycin (PI3K/Akt/mTOR) Signaling. Med Sci Monit 2018; 24:7459-7468. [PMID: 30338764 PMCID: PMC6354638 DOI: 10.12659/msm.908142] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Hypoxic-ischemic brain injury in the perinatal period is a main cause of perinatal mortality and neurologic complications in neonates and children. Recent studies have focused on the neuroprotective effect of anesthetic drugs. The volatile anesthetic isoflurane has been shown to exert neuroprotective effects in cerebral ischemia. Mangiferin is a natural polyphenol with various pharmacological properties, including antioxidant and ant-tumor effects. This study aimed to determine whether mangiferin potentiates the neuroprotective effects of isoflurane and also if mangiferin when administered alone exerts neuroprotective effects following hypoxic-ischemic brain injury. Material/Methods Sprague-Dawley rats were subjected to cerebral hypoxic ischemia on postnatal day 10 (P10). Mangiferin (50, 100, or 200 mg/kg b.w.) was intragastrically administered from P3 to P12 and 1 h prior to insult on the day of ischemic induction. At 3 h after hypoxia-ischemia (HI) insult, separate groups of rat pups were exposed to isoflurane (1.5%) for 6 h. Following 48 h of HI, the rats were sacrificed and brain tissues were used for analysis. Results Mangiferin treatment attenuated neuronal apoptosis and reduced cerebral infarct volume. The expression of cleaved caspase-3 and apoptotic cascade proteins were regulated. The levels of reactive oxygen species (ROS) and malondialdehyde were reduced by mangiferin and/or isoflurane exposure. The levels of antioxidant glutathione were considerably raised under HI injury, which was modulated by mangiferin and isoflurane exposure. The PI3K/Akt signaling pathway, which was downregulated following HI insult, was activated by mangiferin and/or isoflurane. Conclusions This study reveals the potent neuroprotective efficacy of mangiferin against HI-induced brain injury via effectively modulating apoptotic pathways, ROS levels, and PI3K/Akt cascades while potentiating protective effects of isoflurane.
Collapse
Affiliation(s)
- Jia-Shui Xi
- Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Yu-Fen Wang
- Department of Pediatrics, Shandong Provincial Third Hospital, Jinan, Shandong, China (mainland)
| | - Xin-Xin Long
- Department of Pediatrics, Shandong Provincial Third Hospital, Jinan, Shandong, China (mainland)
| | - Yan Ma
- Department of Pediatrics, Shandong Provincial Third Hospital, Jinan, Shandong, China (mainland)
| |
Collapse
|
29
|
Huang S, Yang C, Li M, Wang B, Chen H, Fu D, Chong T. Effect of dual mTOR inhibitor on TGFβ1-induced fibrosis in primary human urethral scar fibroblasts. Biomed Pharmacother 2018; 106:1182-1187. [DOI: 10.1016/j.biopha.2018.07.070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 11/17/2022] Open
|
30
|
Walker NM, Mazzoni SM, Vittal R, Fingar DC, Lama VN. c-Jun N-terminal kinase (JNK)-mediated induction of mSin1 expression and mTORC2 activation in mesenchymal cells during fibrosis. J Biol Chem 2018; 293:17229-17239. [PMID: 30217824 DOI: 10.1074/jbc.ra118.003926] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/06/2018] [Indexed: 02/03/2023] Open
Abstract
Mammalian target of rapamycin complex 2 (mTORC2) has been shown to regulate mTORC1/4E-BP1/eIF4E signaling and collagen I expression in mesenchymal cells (MCs) during fibrotic activation. Here we investigated the regulation of the mTORC2 binding partner mammalian stress-activated protein kinase-interacting protein 1 (mSin1) in MCs derived from human lung allografts and identified a novel role for mSin1 during fibrosis. mSin1 was identified as a common downstream target of key fibrotic pathways, and its expression was increased in MCs in response to pro-fibrotic mediators: lysophosphatidic acid (LPA), transforming growth factor β, and interleukin 13. Fibrotic MCs had higher mSin1 protein levels than nonfibrotic MCs, and siRNA-mediated silencing of mSIN1 inhibited collagen I expression and mTORC1/2 activity in these cells. Autocrine LPA signaling contributed to constitutive up-regulation of mSin1 in fibrotic MCs, and mSin1 was decreased because of LPA receptor 1 siRNA treatment. We identified c-Jun N-terminal kinase (JNK) as a key intermediary in mSin1 up-regulation by the pro-fibrotic mediators, as pharmacological and siRNA-mediated inhibition of JNK prevented the LPA-induced mSin1 increase. Proteasomal inhibition rescued mSin1 levels after JNK inhibition in LPA-treated MCs, and the decrease in mSin1 ubiquitination in response to LPA was counteracted by JNK inhibitors. Constitutive JNK1 overexpression induced mSin1 expression and could drive mTORC2 and mTORC1 activation and collagen I expression in nonfibrotic MCs, effects that were reversed by siRNA-mediated mSIN1 silencing. These results indicate that LPA stabilizes mSin1 protein expression via JNK signaling by blocking its proteasomal degradation and identify the LPA/JNK/mSin1/mTORC/collagen I pathway as critical for fibrotic activation of MCs.
Collapse
Affiliation(s)
- Natalie M Walker
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and
| | - Serina M Mazzoni
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and
| | - Ragini Vittal
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and
| | - Diane C Fingar
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-0360
| | - Vibha N Lama
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and
| |
Collapse
|
31
|
Molina-Molina M, Machahua-Huamani C, Vicens-Zygmunt V, Llatjós R, Escobar I, Sala-Llinas E, Luburich-Hernaiz P, Dorca J, Montes-Worboys A. Anti-fibrotic effects of pirfenidone and rapamycin in primary IPF fibroblasts and human alveolar epithelial cells. BMC Pulm Med 2018; 18:63. [PMID: 29703175 PMCID: PMC5922028 DOI: 10.1186/s12890-018-0626-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 04/16/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Pirfenidone, a pleiotropic anti-fibrotic treatment, has been shown to slow down disease progression of idiopathic pulmonary fibrosis (IPF), a fatal and devastating lung disease. Rapamycin, an inhibitor of fibroblast proliferation could be a potential anti-fibrotic drug to improve the effects of pirfenidone. METHODS Primary lung fibroblasts from IPF patients and human alveolar epithelial cells (A549) were treated in vitro with pirfenidone and rapamycin in the presence or absence of transforming growth factor β1 (TGF-β). Extracellular matrix protein and gene expression of markers involved in lung fibrosis (tenascin-c, fibronectin, collagen I [COL1A1], collagen III [COL3A1] and α-smooth muscle actin [α-SMA]) were analyzed. A cell migration assay in pirfenidone, rapamycin and TGF-β-containing media was performed. RESULTS Gene and protein expression of tenascin-c and fibronectin of fibrotic fibroblasts were reduced by pirfenidone or rapamycin treatment. Pirfenidone-rapamycin treatment did not revert the epithelial to mesenchymal transition pathway activated by TGF-β. However, the drug combination significantly abrogated fibroblast to myofibroblast transition. The inhibitory effect of pirfenidone on fibroblast migration in the scratch-wound assay was potentiated by rapamycin combination. CONCLUSIONS These findings indicate that the combination of pirfenidone and rapamycin widen the inhibition range of fibrogenic markers and prevents fibroblast migration. These results would open a new line of research for an anti-fibrotic combination therapeutic approach.
Collapse
Affiliation(s)
- M. Molina-Molina
- Department of Pneumology, Bellvitge University Hospital, Barcelona, Spain
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), ISCIII, Madrid, Spain
| | - C. Machahua-Huamani
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
| | - V. Vicens-Zygmunt
- Department of Pneumology, Bellvitge University Hospital, Barcelona, Spain
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
| | - R. Llatjós
- Department of Pathology, Bellvitge University Hospital, Barcelona, Spain
| | - I. Escobar
- Department of Thoracic Surgery, Bellvitge University Hospital, Barcelona, Spain
| | - E. Sala-Llinas
- Research Network in Respiratory Diseases (CIBERES), ISCIII, Madrid, Spain
- Department of Penumology, Son Espases University Hospital, Palma de Mallorca, Spain
| | - P. Luburich-Hernaiz
- Servei de Diagnostic per la Imatge El Prat (SDPI El Prat) Department of Radiology, Bellvitge University Hospital, Barcelona, Spain
| | - J. Dorca
- Department of Pneumology, Bellvitge University Hospital, Barcelona, Spain
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), ISCIII, Madrid, Spain
| | - A. Montes-Worboys
- Department of Pneumology, Bellvitge University Hospital, Barcelona, Spain
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), ISCIII, Madrid, Spain
- Laboratori de Pneumologia Experimental (Lab. 4126). IDIBELL, Pavelló de Govern. Campus de Bellvitge, Universitat de Barcelona, Hospital de Bellvitge, Carrer de la Feixa Llarga, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
32
|
Caja L, Dituri F, Mancarella S, Caballero-Diaz D, Moustakas A, Giannelli G, Fabregat I. TGF-β and the Tissue Microenvironment: Relevance in Fibrosis and Cancer. Int J Mol Sci 2018. [PMID: 29701666 DOI: 10.3390/ijms19051294.pmid:29701666;pmcid:pmc5983604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a cytokine essential for the induction of the fibrotic response and for the activation of the cancer stroma. Strong evidence suggests that a strong cross-talk exists among TGF-β and the tissue extracellular matrix components. TGF-β is stored in the matrix as part of a large latent complex bound to the latent TGF-β binding protein (LTBP) and matrix binding of latent TGF-β complexes, which is required for an adequate TGF-β function. Once TGF-β is activated, it regulates extracellular matrix remodelling and promotes a fibroblast to myofibroblast transition, which is essential in fibrotic processes. This cytokine also acts on other cell types present in the fibrotic and tumour microenvironment, such as epithelial, endothelial cells or macrophages and it contributes to the cancer-associated fibroblast (CAF) phenotype. Furthermore, TGF-β exerts anti-tumour activity by inhibiting the host tumour immunosurveillance. Aim of this review is to update how TGF-β and the tissue microenvironment cooperate to promote the pleiotropic actions that regulate cell responses of different cell types, essential for the development of fibrosis and tumour progression. We discuss recent evidences suggesting the use of TGF-β chemical inhibitors as a new line of defence against fibrotic disorders or cancer.
Collapse
Affiliation(s)
- Laia Caja
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Francesco Dituri
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Serena Mancarella
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Daniel Caballero-Diaz
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Aristidis Moustakas
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, L'Hospitalet, 08907 Barcelona, Spain.
| |
Collapse
|
33
|
Caja L, Dituri F, Mancarella S, Caballero-Diaz D, Moustakas A, Giannelli G, Fabregat I. TGF-β and the Tissue Microenvironment: Relevance in Fibrosis and Cancer. Int J Mol Sci 2018; 19:ijms19051294. [PMID: 29701666 PMCID: PMC5983604 DOI: 10.3390/ijms19051294] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a cytokine essential for the induction of the fibrotic response and for the activation of the cancer stroma. Strong evidence suggests that a strong cross-talk exists among TGF-β and the tissue extracellular matrix components. TGF-β is stored in the matrix as part of a large latent complex bound to the latent TGF-β binding protein (LTBP) and matrix binding of latent TGF-β complexes, which is required for an adequate TGF-β function. Once TGF-β is activated, it regulates extracellular matrix remodelling and promotes a fibroblast to myofibroblast transition, which is essential in fibrotic processes. This cytokine also acts on other cell types present in the fibrotic and tumour microenvironment, such as epithelial, endothelial cells or macrophages and it contributes to the cancer-associated fibroblast (CAF) phenotype. Furthermore, TGF-β exerts anti-tumour activity by inhibiting the host tumour immunosurveillance. Aim of this review is to update how TGF-β and the tissue microenvironment cooperate to promote the pleiotropic actions that regulate cell responses of different cell types, essential for the development of fibrosis and tumour progression. We discuss recent evidences suggesting the use of TGF-β chemical inhibitors as a new line of defence against fibrotic disorders or cancer.
Collapse
Affiliation(s)
- Laia Caja
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Francesco Dituri
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Serena Mancarella
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Daniel Caballero-Diaz
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Aristidis Moustakas
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, L'Hospitalet, 08907 Barcelona, Spain.
| |
Collapse
|
34
|
Gokey JJ, Sridharan A, Xu Y, Green J, Carraro G, Stripp BR, Perl AKT, Whitsett JA. Active epithelial Hippo signaling in idiopathic pulmonary fibrosis. JCI Insight 2018; 3:98738. [PMID: 29563341 DOI: 10.1172/jci.insight.98738] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/13/2018] [Indexed: 12/21/2022] Open
Abstract
Hippo/YAP signaling plays pleiotropic roles in the regulation of cell proliferation and differentiation during organogenesis and tissue repair. Herein we demonstrate increased YAP activity in respiratory epithelial cells in lungs of patients with idiopathic pulmonary fibrosis (IPF), a common, lethal form of interstitial lung disease (ILD). Immunofluorescence staining in IPF epithelial cells demonstrated increased nuclear YAP and loss of MST1/2. Bioinformatic analyses of epithelial cell RNA profiles predicted increased activity of YAP and increased canonical mTOR/PI3K/AKT signaling in IPF. Phospho-S6 (p-S6) and p-PTEN were increased in IPF epithelial cells, consistent with activation of mTOR signaling. Expression of YAP (S127A), a constitutively active form of YAP, in human bronchial epithelial cells (HBEC3s) increased p-S6 and p-PI3K, cell proliferation and migration, processes that were inhibited by the YAP-TEAD inhibitor verteporfin. Activation of p-S6 was required for enhancing and stabilizing YAP, and the p-S6 inhibitor temsirolimus blocked nuclear YAP localization and suppressed expression of YAP target genes CTGF, AXL, and AJUBA (JUB). YAP and mTOR/p-S6 signaling pathways interact to induce cell proliferation and migration, and inhibit epithelial cell differentiation that may contribute to the pathogenesis of IPF.
Collapse
Affiliation(s)
- Jason J Gokey
- Division of Neonatology, Perinatal and Pulmonary Biology, and
| | | | - Yan Xu
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jenna Green
- Division of Neonatology, Perinatal and Pulmonary Biology, and
| | - Gianni Carraro
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Barry R Stripp
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, USA
| | | | | |
Collapse
|
35
|
Abstract
The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR)-dependent pathway is one of the most integral pathways linked to cell metabolism, proliferation, differentiation, and survival. This pathway is dysregulated in a variety of diseases, including neoplasia, immune-mediated diseases, and fibroproliferative diseases such as pulmonary fibrosis. The mTOR kinase is frequently referred to as the master regulator of this pathway. Alterations in mTOR signaling are closely associated with dysregulation of autophagy, inflammation, and cell growth and survival, leading to the development of lung fibrosis. Inhibitors of mTOR have been widely studied in cancer therapy, as they may sensitize cancer cells to radiation therapy. Studies also suggest that mTOR inhibitors are promising modulators of fibroproliferative diseases such as idiopathic pulmonary fibrosis (IPF) and radiation-induced pulmonary fibrosis (RIPF). Therefore, mTOR represents an attractive and unique therapeutic target in pulmonary fibrosis. In this review, we discuss the pathological role of mTOR kinase in pulmonary fibrosis and examine how mTOR inhibitors may mitigate fibrotic progression.
Collapse
|
36
|
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway has been studied in the context of an impressive number of biological processes and disease states, including major diseases of the lung such as idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease, as well as the rare condition lymphangioleiomyomatosis. The involvement of mTOR in so many disease states (in and out of the lung) raises the question how one signaling pathway can have overlapping but diverse roles seemingly everywhere. Findings in the last decade have placed the mTOR pathway in a new context as an important, conserved mediator of the aging process. This offers one explanation for the pleiotropic effects of mTOR: -that many chronic diseases are also diseases of aging and that pathways modulating aging will have widespread effects on associated disease. However, this may not be the entire story, because mTOR is also implicated in a large number of diseases not linked to aging. In this article, we discuss the current state of knowledge regarding mTOR, especially in the context of lung pathologies, and offer a potential explanation for its widespread involvement in human disease.
Collapse
|
37
|
Rashidi M, Bandala-Sanchez E, Lawlor KE, Zhang Y, Neale AM, Vijayaraj SL, O'Donoghue R, Wentworth JM, Adams TE, Vince JE, Harrison LC. CD52 inhibits Toll-like receptor activation of NF-κB and triggers apoptosis to suppress inflammation. Cell Death Differ 2017; 25:392-405. [PMID: 29244050 DOI: 10.1038/cdd.2017.173] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/29/2017] [Accepted: 09/13/2017] [Indexed: 12/11/2022] Open
Abstract
Soluble CD52 is a small glycoprotein that suppresses T-cell activation, but its effect on innate immune cell function is unknown. Here we demonstrate that soluble CD52 inhibits Toll-like receptor and tumor necrosis factor receptor signaling to limit activation of NF-κB and thereby suppress the production of inflammatory cytokines by macrophages, monocytes and dendritic cells. At higher concentrations, soluble CD52 depletes the short-lived pro-survival protein MCL-1, contributing to activation of the BH3-only proteins BAX and BAK to cause intrinsic apoptotic cell death. In vivo, administration of soluble CD52 suppresses lipopolysaccharide (LPS)-induced cytokine secretion and other features of endotoxic shock, whereas genetic deletion of CD52 exacerbates LPS responses. Thus, soluble CD52 exhibits broad immune suppressive effects that signify its potential as an immunotherapeutic agent.
Collapse
Affiliation(s)
- Maryam Rashidi
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Esther Bandala-Sanchez
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Kate E Lawlor
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yuxia Zhang
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alana M Neale
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Swarna L Vijayaraj
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Robert O'Donoghue
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - John M Wentworth
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Timothy E Adams
- CSIRO Manufacturing and Node of the National Biologics Facility, Parkville, Victoria 3052, Australia
| | - James E Vince
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Leonard C Harrison
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
38
|
Zhou L, Liu S, Han M, Ma Y, Feng S, Zhao J, Lu H, Yuan X, Cheng J. miR-185 Inhibits Fibrogenic Activation of Hepatic Stellate Cells and Prevents Liver Fibrosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 10:91-102. [PMID: 29499960 PMCID: PMC5735261 DOI: 10.1016/j.omtn.2017.11.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 02/07/2023]
Abstract
Recent studies have shown the effect of microRNAs on HSC activation and transformation, which is essential for the pathogenesis of liver fibrosis. In our study, we explored the role of miR-185 in liver fibrosis. Plasma miR-185 was detected in hepatitis B virus-related liver fibrosis patients (S2/3, n = 10) by Illumina HiSeq sequencing, and healthy volunteers were selected (n = 8) as the control group. We found that the plasma miR-185 level in fibrosis patients was significantly downregulated. CCl4-induced fibrosis tissues in mouse livers and TGF-β1-activated HSCs also presented downregulated miR-185 concomitant with an increased expression of RHEB and RICTOR. To explore the correlations, LX-2 cells were transiently transfected with miR-185 mimics. The expression levels of α-SMA, collagen I, and collagen III were decreased as well as RHEB and RICTOR. Inhibition of endogenous miR-185 increased fibrogenic activity. Furthermore, dual-luciferase reporter assays indicated that miR-185 inhibited the expression of RHEB and RICTOR by directly targeting their 3' UTRs. Moreover, silencing RHEB and RICTOR suppressed α-SMA and collagen expression levels. In conclusion, miR-185 prevents liver fibrogenesis by inhibiting HSC activation via inhibition of RHEB and RICTOR. These results provide new insights into the mechanisms behind the anti-fibrotic effect of miR-185.
Collapse
Affiliation(s)
- Li Zhou
- Peking University Ditan Teaching Hospital, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China; Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Shunai Liu
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China; Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Ming Han
- Peking University Ditan Teaching Hospital, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China; Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yanhua Ma
- Peking University Ditan Teaching Hospital, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Shenghu Feng
- Peking University Ditan Teaching Hospital, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Jing Zhao
- Peking University Ditan Teaching Hospital, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Hongping Lu
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China; Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xiaoxue Yuan
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China; Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Jun Cheng
- Peking University Ditan Teaching Hospital, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China; Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| |
Collapse
|
39
|
Hao X, Du Y, Qian L, Li D, Liu X. Upregulation of long noncoding RNA AP003419.16 predicts high risk of aging‑associated idiopathic pulmonary fibrosis. Mol Med Rep 2017; 16:8085-8091. [PMID: 28944926 PMCID: PMC5779893 DOI: 10.3892/mmr.2017.7607] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 07/03/2017] [Indexed: 01/03/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are able to regulate adjacent genes and thus participate in the incidence in the present study has identified lncRNA AP003419.16, adjacent to the protein-coding gene ribosomal protein S6 kinase B-2 (RPS6KB2). RPS6KB2 is believed to be involved in the process of aging and idiopathic pulmonary fibrosis (IPF), due to its activation by growth factors and regulation by the protein kinase mTOR signaling pathway. The results of the present study indicated that the expression of AP003419.16 increased significantly in patients with IPF, whereas its adjacent gene ribosomal protein S6 kinase B-2 increased simultaneously. AP003419.16 expression may be used to predict an increased risk of aging-associated IPF. The present study provided a molecular hypothesis of IPF occurrence in the aging process, in addition to novel molecular targets for the clinical treatment of IPF.
Collapse
Affiliation(s)
- Xiaoyan Hao
- Department of Geriatrics, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yufeng Du
- Department of Geriatrics, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Li Qian
- Department of Geriatrics, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Dan Li
- Department of Geriatrics, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xuejun Liu
- Department of Geriatrics, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
40
|
Yue H, Zhao Y, Wang H, Ma F, Liu F, Shen S, Hou Y, Dou H. Anti-fibrosis effect for Hirsutella sinensis mycelium based on inhibition of mTOR p70S6K phosphorylation. Innate Immun 2017; 23:615-624. [DOI: 10.1177/1753425917726361] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hirsutella sinensis, cultured in vitro, is an attractive substitute for Cordyceps sinensis as health supplement. The aim of this study was to demonstrate whether H. sinensis mycelium (HSM) attenuates murine pulmonary fibrosis induced by bleomycin and to explore the underlying molecular mechanisms. Using lung fibrosis modle induced by intratracheal instillation of bleomycin (BLM; 4 mg/kg), we observed that the administration of HSM reduced HYP, TGF-β1 and the production of several pro-fibrosis cytokines (α-smooth muscle actin, fibronectin and vimentin) in fibrotic mice lung sections. Histopathological examination of lung tissues also demonstrated that HSM improved BLM-induced pathological damage. Concurrently, HSM supplementation markedly reduced the chemotaxis of alveolar macrophages and potently suppressed the expression of inflammatory cytokines. Also, HSM influenced Th1/Th2 and Th17/Treg imbalance and blocked the phosphorylation of mTOR pathway in vivo. Alveolar epithelial A549 cells acquired a mesenchymal phenotype and an increased expression of myofibroblast markers of differentiation (vimentin and fibronectin) after treatment with TGF-β1. HSM suppressed these markers and blocked the phosphorylation of mTOR pathway in vitro. The results provide evidence supporting the use of HSM in the intervention of pulmonary fibrosis and suggest that HSM is a potential therapeutic agent for lung fibrosis.
Collapse
Affiliation(s)
- Huimin Yue
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Yarong Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Haining Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Feiya Ma
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Fei Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Sunan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
41
|
Expression of mTOR in Primary Pterygium and its Correlation with α-Smooth Muscle Actin. Eur J Ophthalmol 2017. [PMID: 28623647 DOI: 10.5301/ejo.5000985] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Purpose Mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that has been shown to affect many cellular functions, such as cell growth, proliferation, and metabolism. However, there has been minimal focus on the expression of mTOR in pterygium. The purpose of this study was to investigate the expression of mTOR and the correlation between the levels of mTOR and α-smooth muscle actin (α-SMA, a marker of transdifferentiation) in pterygium. Methods Primary pterygium samples from 28 patients and normal conjunctival samples from 16 patients were surgically removed and analyzed. The expression levels of mTOR and α-SMA in the excised specimens were assessed using immunohistochemistry and Western blotting. Furthermore, correlations between the mTOR and α-SMA expression levels were studied. Results The expression of mTOR and α-SMA was significantly higher in the pterygium tissues than in normal conjunctiva tissues. A significant positive correlation was detected between the number of mTOR-immunopositive fibroblasts and the number of α-SMA-immunopositive fibroblasts (ρ = 0.463, p = 0.0078). Additionally, mTOR expression was significantly correlated with α-SMA expression (ρ = 0.269, p = 0.031) in pterygium. Conclusions There was an increased expression of mTOR in pterygium samples compared to that in normal conjunctival tissues, with a positive correlation with α-SMA expression. These findings might be involved in the pathogenesis of pterygium.
Collapse
|
42
|
XPLN is modulated by HDAC inhibitors and negatively regulates SPARC expression by targeting mTORC2 in human lung fibroblasts. Pulm Pharmacol Ther 2017; 44:61-69. [DOI: 10.1016/j.pupt.2017.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/12/2017] [Indexed: 11/19/2022]
|
43
|
Nobiletin improves propofol-induced neuroprotection via regulating Akt/mTOR and TLR 4/NF-κB signaling in ischemic brain injury in rats. Biomed Pharmacother 2017; 91:494-503. [PMID: 28478273 DOI: 10.1016/j.biopha.2017.04.048] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 04/09/2017] [Accepted: 04/10/2017] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Stroke is regarded as one of the main health concerns globally, presenting with high mortality and morbidity rates. Cerebral ischemic damage and infarction are critically associated with stroke. Various mechanisms related to inflammation, oxidative stress and excitotoxicity are found to be involved in ischemic damage. Very short time period for treatment has necessitated in development of more effective neuroprotective agents. Study aimed in investigated the effects of nobiletin on experimentally induced ischemic brain injury and also to assess whether nobiletin potentiated the neuroprotective effects of propofol. METHODS Male Sprague-Dawley rats were subjected to ischemia/reperfusion (I/R) injury. Induction of cerebral infarction and I/R was done by middle cerebral artery occlusion (MCAO). Nobiletin (100 or 200mg/kg b.wt.) was intragastrically administered to rats for 9 days before ischemia induction and on the day of induction nobiletin was administered an hour prior. Separate group of rats were post-conditioned with propofol (50mg/kg/h; i.v.) for 30min following 24h of reperfusion. RESULTS Propofol post-conditioning either with or without administration of nobiletin prior I/R injury attenuated pulmonary edema, neuronal apoptosis and reduced cerebral infarct volume. Overproduction of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) and nitric oxide following I/R were reduced. Propofol either alone or with prior nobiletin treatment had down-regulated TLR4 and TLR4-mediated NF-κB signaling and caused activation of Akt/mTOR cascade. CONCLUSION Propofol post-conditioning either with nobiletin prior I/R injury was found to be more effective than propofol alone, suggesting the positive effects of nobiletin on propofol-mediated anti-inflammatory and neuroprotective effects.
Collapse
|
44
|
Ferguson KT, Torr EE, Bernau K, Leet J, Sherris D, Sandbo N. The Novel mTOR Complex 1/2 Inhibitor P529 Inhibits Human Lung Myofibroblast Differentiation. J Cell Biochem 2017; 118:2241-2249. [PMID: 28078713 DOI: 10.1002/jcb.25878] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/10/2017] [Indexed: 12/19/2022]
Abstract
Idiopathic pulmonary fibrosis is a progressive and deadly disorder with very few therapeutic options. Palomid 529 (8-(1-hydroxyethyl)-2-methoxy-3-(4-methoxybenzyloxy)-benzo[c]chromen-6-one; P529) is a novel dual inhibitor of mechanistic target of rapamycin complex 1/2 (mTORC1/2). In these studies, we investigated the effect of P529 on TGF-β-dependent signaling and myofibroblast differentiation. TGF-β-induced phosphorylation of the mTORC1 targets, p70 S6 kinase 1 (S6K1), and eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), were both dose dependently inhibited by P529 in human lung fibroblasts with maximal inhibition occurring between 10 and 20 μM. mTORC2-mediated phosphorylation of Akt at the S473 site was partially inhibited with a similar dose dependency, as was TGF-β-induced myofibroblast differentiation. Protein levels of TGF-β-induced fibronectin and collagen were similarly decreased by P529. At this dose, there was also inhibition of mRNA transcript levels for Col1 and α-SMA, suggesting inhibition of transcriptional activation. However, there was no effect of P529 on canonical TGF-β-induced Smad signaling, as assessed by receptor-associated Smad2/3 phosphorylation, Smad2/3/4 translocation, or Smad-driven gene expression, as assessed by Smad-binding element driven luciferase. Conversely, activation of mTORC1/2 signaling was dependent on TGF-β type I receptor (ALK5) signaling and on Smad2/3 expression. P529 treatment disrupted TGF-β-induced actin stress fiber formation during myofibroblast differentiation, the deposition of new extracellular fibronectin matrix, and linear wound closure by fibroblasts. Likewise, mTOR knockdown inhibited TGF-β-induced myofibroblast differentiation. In conclusion, P529 inhibits TGF-β-induced myofibroblast differentiation, actin stress fiber formation, and matrix protein expression and deposition. Inhibition of mTORC1/2 by P529 may be a promising approach to inhibit in vivo fibrosis. J. Cell. Biochem. 118: 2241-2249, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Keith T Ferguson
- Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 600 Highland Ave, Madison, Wisconsin, 53792
| | - Elizabeth E Torr
- Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 600 Highland Ave, Madison, Wisconsin, 53792
| | - Ksenija Bernau
- Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 600 Highland Ave, Madison, Wisconsin, 53792
| | - Jonathan Leet
- Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 600 Highland Ave, Madison, Wisconsin, 53792
| | - David Sherris
- GenAdam Therapeutics, Inc, 37 Neillian Crescent, Jamaica Plain, Massachusetts, 02130
| | - Nathan Sandbo
- Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 600 Highland Ave, Madison, Wisconsin, 53792
| |
Collapse
|
45
|
Chung EJ, Sowers A, Thetford A, McKay-Corkum G, Chung SI, Mitchell JB, Citrin DE. Mammalian Target of Rapamycin Inhibition With Rapamycin Mitigates Radiation-Induced Pulmonary Fibrosis in a Murine Model. Int J Radiat Oncol Biol Phys 2016; 96:857-866. [PMID: 27663762 DOI: 10.1016/j.ijrobp.2016.07.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/18/2016] [Accepted: 07/21/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Radiation-induced pulmonary fibrosis (RIPF) is a late toxicity of therapeutic radiation. Signaling of the mammalian target of rapamycin drives several processes implicated in RIPF, including inflammatory cytokine production, fibroblast proliferation, and epithelial senescence. We sought to determine if mammalian target of rapamycin inhibition with rapamycin would mitigate RIPF. METHODS AND MATERIALS C57BL/6NCr mice received a diet formulated with rapamycin (14 mg/kg food) or a control diet 2 days before and continuing for 16 weeks after exposure to 5 daily fractions of 6 Gy of thoracic irradiation. Fibrosis was assessed with Masson trichrome staining and hydroxyproline assay. Cytokine expression was evaluated by quantitative real-time polymerase chain reaction. Senescence was assessed by staining for β-galactosidase activity. RESULTS Administration of rapamycin extended the median survival of irradiated mice compared with the control diet from 116 days to 156 days (P=.006, log-rank test). Treatment with rapamycin reduced hydroxyproline content compared with the control diet (irradiation plus vehicle, 45.9 ± 11.8 μg per lung; irradiation plus rapamycin, 21.4 ± 6.0 μg per lung; P=.001) and reduced visible fibrotic foci. Rapamycin treatment attenuated interleukin 1β and transforming growth factor β induction in irradiated lungs compared with the control diet. Type II pneumocyte senescence after irradiation was reduced with rapamycin treatment at 16 weeks (3-fold reduction at 16 weeks, P<.001). CONCLUSIONS Rapamycin protected against RIPF in a murine model. Rapamycin treatment reduced inflammatory cytokine expression, extracellular matrix production, and senescence in type II pneumocytes.
Collapse
Affiliation(s)
- Eun Joo Chung
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Anastasia Sowers
- Radiation Biology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Angela Thetford
- Radiation Biology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Grace McKay-Corkum
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Su I Chung
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Deborah E Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
46
|
Rictor/mTORC2 signaling mediates TGFβ1-induced fibroblast activation and kidney fibrosis. Kidney Int 2015; 88:515-27. [PMID: 25970154 PMCID: PMC4558569 DOI: 10.1038/ki.2015.119] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 02/23/2015] [Accepted: 02/26/2015] [Indexed: 02/06/2023]
Abstract
The mammalian target of rapamycin (mTOR) was recently identified in two structurally distinct multiprotein complexes: mTORC1 and mTORC2. Previously, we found that Rictor/mTORC2 protects against cisplatin-induced acute kidney injury, but the role and mechanisms for Rictor/mTORC2 in TGFβ1-induced fibroblast activation and kidney fibrosis remains unknown. To study this, we initially treated NRK-49F cells with TGFβ1 and found that TGFβ1 could activate Rictor/mTORC2 signaling in cultured cells. Blocking Rictor/mTORC2 signaling with Rictor or Akt1 small interfering RNAs markedly inhibited TGFβ1-induced fibronection and α-smooth muscle actin expression. Ensuing western blotting or immunostaining results showed that Rictor/mTORC2 signaling was activated in kidney interstitial myofibroblasts from mice with unilateral ureteral obstruction. Next, a mouse model with fibroblast-specific deletion of Rictor was generated. These knockout mice were normal at birth and had no obvious kidney dysfunction or kidney morphological abnormality within 2 months of birth. Compared with control littermates, the kidneys of Rictor knockout mice developed less interstitial extracellular matrix deposition and inflammatory cell infiltration at 1 or 2 weeks after ureteral obstruction. Thus our study suggests that Rictor/mTORC2 signaling activation mediates TGFβ1-induced fibroblast activation and contributes to the development of kidney fibrosis. This may provide a therapeutic target for chronic kidney diseases.
Collapse
|
47
|
PI3K/Akt signaling in osteosarcoma. Clin Chim Acta 2015; 444:182-92. [PMID: 25704303 DOI: 10.1016/j.cca.2014.12.041] [Citation(s) in RCA: 249] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/24/2014] [Accepted: 12/09/2014] [Indexed: 12/19/2022]
Abstract
Osteosarcoma (OS) is the most common nonhematologic bone malignancy in children and adolescents. Despite the advances of adjuvant chemotherapy and significant improvement of survival, the prognosis remains generally poor. As such, the search for more effective anti-OS agents is urgent. The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is thought to be one of the most important oncogenic pathways in human cancer. An increasing body of evidence has shown that this pathway is frequently hyperactivated in OS and contributes to disease initiation and development, including tumorigenesis, proliferation, invasion, cell cycle progression, inhibition of apoptosis, angiogenesis, metastasis and chemoresistance. Inhibition of this pathway through small molecule compounds represents an attractive potential therapeutic approach for OS. The aim of this review is to summarize the roles of the PI3K/Akt pathway in the development and progression of OS, and to highlight the therapeutic potential of targeting this signaling pathway. Knowledge obtained from the application of these compounds will help in further understanding the pathogenesis of OS and designing subsequent treatment strategies.
Collapse
|