1
|
Cui T, Huang Z, Luo K, Nie J, Xv Y, Zeng Z, Liao L, Yang X, Zhou H. Identification of Hub Genes and Prediction of Targeted Drugs for Rheumatoid Arthritis and Idiopathic Pulmonary Fibrosis. Biochem Genet 2024; 62:5157-5178. [PMID: 38334875 DOI: 10.1007/s10528-023-10650-z] [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: 06/25/2023] [Accepted: 12/25/2023] [Indexed: 02/10/2024]
Abstract
There is a potential link between rheumatoid arthritis (RA) and idiopathic pulmonary fibrosis (IPF). The aim of this study is to investigate the molecular processes that underlie the development of these two conditions by bioinformatics methods. The gene expression samples for RA (GSE77298) and IPF (GSE24206) were retrieved from the Gene Expression Omnibus (GEO) database. After identifying the overlapping differentially expressed genes (DEGs) for RA and IPF, we conducted functional annotation, protein-protein interaction (PPI) network analysis, and hub gene identification. Finally, we used the hub genes to predict potential medications for the treatment of both disorders. We identified 74 common DEGs for further analysis. Functional analysis demonstrated that cellular components, biological processes, and molecular functions all played a role in the emergence and progression of RA and IPF. Using the cytoHubba plugin, we identified 7 important hub genes, namely COL3A1, SDC1, CCL5, CXCL13, MMP1, THY1, and BDNF. As diagnostic indicators for RA, SDC1, CCL5, CXCL13, MMP1, and THY1 showed favorable values. For IPF, COL3A1, SDC1, CCL5, CXCL13, THY1, and BDNF were favorable diagnostic markers. Furthermore, we predicted 61 Chinese and 69 Western medications using the hub genes. Our research findings demonstrate a shared pathophysiology between RA and IPF, which may provide new insights for more mechanistic research and more effective treatments. These common pathways and hub genes identified in our study offer potential opportunities for developing more targeted therapies that can address both disorders.
Collapse
Affiliation(s)
- Ting Cui
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of TCM, Chengdu, 610000, Sichuan, China
| | - Zhican Huang
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of TCM, Chengdu, 610000, Sichuan, China
| | - Kun Luo
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of TCM, Chengdu, 610000, Sichuan, China
| | - Jingwei Nie
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of TCM, Chengdu, 610000, Sichuan, China
| | - Yimei Xv
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of TCM, Chengdu, 610000, Sichuan, China
| | - Zhu Zeng
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of TCM, Chengdu, 610000, Sichuan, China
| | - Linghan Liao
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of TCM, Chengdu, 610000, Sichuan, China
| | - Xin Yang
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of TCM, Chengdu, 610000, Sichuan, China
| | - Haiyan Zhou
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of TCM, Chengdu, 610000, Sichuan, China.
| |
Collapse
|
2
|
Singh A, Mommers-Elshof ETAM, Vijver SV, Jansen JHM, Gonder S, Lebbink RJ, Bihan D, Farndale RW, Boon L, Langermann S, Leusen JHW, Flies D, Meyaard L, Pascoal Ramos MI. Leukocyte-associated immunoglobulin-like receptor-1 blockade in combination with programmed death-ligand 1 targeting therapy mediates increased tumour control in mice. Cancer Immunol Immunother 2024; 73:16. [PMID: 38236251 PMCID: PMC10796629 DOI: 10.1007/s00262-023-03600-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/15/2023] [Indexed: 01/19/2024]
Abstract
Collagen expression and structure in the tumour microenvironment are associated with tumour development and therapy response. Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is a widely expressed inhibitory collagen receptor. LAIR-2 is a soluble homologue of LAIR-1 that competes for collagen binding. Multiple studies in mice implicate blockade of LAIR-1:collagen interaction in cancer as a promising therapeutic strategy. Here, we investigated the role of LAIR-1 in anti-tumour responses. We show that although LAIR-1 inhibits activation, proliferation, and cytokine production of mouse T cells in vitro, tumour outgrowth in LAIR-1-deficient mice did not differ from wild type mice in several in vivo tumour models. Furthermore, treatment with NC410, a LAIR-2-Fc fusion protein, did not result in increased tumour clearance in tested immunocompetent mice, which contrasts with previous data in humanized mouse models. This discrepancy may be explained by our finding that NC410 blocks human LAIR-1:collagen interaction more effectively than mouse LAIR-1:collagen interaction. Despite the lack of therapeutic impact of NC410 monotherapy, mice treated with a combination of NC410 and anti-programmed death-ligand 1 did show reduced tumour burden and increased survival. Using LAIR-1-deficient mice, we showed that this effect seemed to be dependent on the presence of LAIR-1. Taken together, our data demonstrate that the absence of LAIR-1 signalling alone is not sufficient to control tumour growth in multiple immunocompetent mouse models. However, combined targeting of LAIR-1 and PD-L1 results in increased tumour control. Thus, additional targeting of the LAIR-1:collagen pathway with NC410 is a promising approach to treating tumours where conventional immunotherapy is ineffective.
Collapse
Affiliation(s)
- Akashdip Singh
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Eline T A M Mommers-Elshof
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Saskia V Vijver
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - J H Marco Jansen
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
| | - Susanne Gonder
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
| | - Robert Jan Lebbink
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
| | - Dominique Bihan
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | | | - Jeanette H W Leusen
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
| | | | - Linde Meyaard
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - M Ines Pascoal Ramos
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Doherty EL, Aw WY, Warren EC, Hockenberry M, Whitworth CP, Krohn G, Howell S, Diekman BO, Legant WR, Nia HT, Hickey AJ, Polacheck WJ. Patient-derived extracellular matrix demonstrates role of COL3A1 in blood vessel mechanics. Acta Biomater 2023; 166:346-359. [PMID: 37187299 PMCID: PMC10330735 DOI: 10.1016/j.actbio.2023.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023]
Abstract
Vascular Ehlers-Danlos Syndrome (vEDS) is a rare autosomal dominant disease caused by mutations in the COL3A1 gene, which renders patients susceptible to aneurysm and arterial dissection and rupture. To determine the role of COL3A1 variants in the biochemical and biophysical properties of human arterial ECM, we developed a method for synthesizing ECM directly from vEDS donor fibroblasts. We found that the protein content of the ECM generated from vEDS donor fibroblasts differed significantly from ECM from healthy donors, including upregulation of collagen subtypes and other proteins related to ECM structural integrity. We further found that ECM generated from a donor with a glycine substitution mutation was characterized by increased glycosaminoglycan content and unique viscoelastic mechanical properties, including increased time constant for stress relaxation, resulting in a decrease in migratory speed of human aortic endothelial cells when seeded on the ECM. Collectively, these results demonstrate that vEDS patient-derived fibroblasts harboring COL3A1 mutations synthesize ECM that differs in composition, structure, and mechanical properties from healthy donors. These results further suggest that ECM mechanical properties could serve as a prognostic indicator for patients with vEDS, and the insights provided by the approach demonstrate the broader utility of cell-derived ECM in disease modeling. STATEMENT OF SIGNIFICANCE: The role of collagen III ECM mechanics remains unclear, despite reported roles in diseases including fibrosis and cancer. Here, we generate fibrous, collagen-rich ECM from primary donor cells from patients with vascular Ehlers-Danlos syndrome (vEDS), a disease caused by mutations in the gene that encodes collagen III. We observe that ECM grown from vEDS patients is characterized by unique mechanical signatures, including altered viscoelastic properties. By quantifying the structural, biochemical, and mechanical properties of patient-derived ECM, we identify potential drug targets for vEDS, while defining a role for collagen III in ECM mechanics more broadly. Furthermore, the structure/function relationships of collagen III in ECM assembly and mechanics will inform the design of substrates for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Elizabeth L Doherty
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily C Warren
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Max Hockenberry
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Chloe P Whitworth
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Grace Krohn
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Stefanie Howell
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brian O Diekman
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Wesley R Legant
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Hadi Tavakoli Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Anthony J Hickey
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA; McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Bosáková V, De Zuani M, Sládková L, Garlíková Z, Jose SS, Zelante T, Hortová Kohoutková M, Frič J. Lung Organoids—The Ultimate Tool to Dissect Pulmonary Diseases? Front Cell Dev Biol 2022; 10:899368. [PMID: 35912110 PMCID: PMC9326165 DOI: 10.3389/fcell.2022.899368] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/24/2022] [Indexed: 11/15/2022] Open
Abstract
Organoids are complex multicellular three-dimensional (3D) in vitro models that are designed to allow accurate studies of the molecular processes and pathologies of human organs. Organoids can be derived from a variety of cell types, such as human primary progenitor cells, pluripotent stem cells, or tumor-derived cells and can be co-cultured with immune or microbial cells to further mimic the tissue niche. Here, we focus on the development of 3D lung organoids and their use as disease models and drug screening tools. We introduce the various experimental approaches used to model complex human diseases and analyze their advantages and disadvantages. We also discuss validation of the organoids and their physiological relevance to the study of lung diseases. Furthermore, we summarize the current use of lung organoids as models of host-pathogen interactions and human lung diseases such as cystic fibrosis, chronic obstructive pulmonary disease, or SARS-CoV-2 infection. Moreover, we discuss the use of lung organoids derived from tumor cells as lung cancer models and their application in personalized cancer medicine research. Finally, we outline the future of research in the field of human induced pluripotent stem cell-derived organoids.
Collapse
Affiliation(s)
- Veronika Bosáková
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marco De Zuani
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Lucie Sládková
- Institute of Hematology and Blood Transfusion, Prague, Czechia
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
| | - Zuzana Garlíková
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Shyam Sushama Jose
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Teresa Zelante
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Jan Frič
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
- Institute of Hematology and Blood Transfusion, Prague, Czechia
- *Correspondence: Jan Frič,
| |
Collapse
|
5
|
Gracioso Martins AM, Biehl A, Sze D, Freytes DO. Bioreactors for Vocal Fold Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:182-205. [PMID: 33446061 PMCID: PMC8892964 DOI: 10.1089/ten.teb.2020.0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/14/2021] [Indexed: 02/03/2023]
Abstract
It is estimated that almost one-third of the United States population will be affected by a vocal fold (VF) disorder during their lifespan. Promising therapies to treat VF injury and scarring are mostly centered on VF tissue engineering strategies such as the injection of engineered biomaterials and cell therapy. VF tissue engineering, however, is a challenging field as the biomechanical properties, structure, and composition of the VF tissue change upon exposure to mechanical stimulation. As a result, the development of long-term VF treatment strategies relies on the characterization of engineered tissues under a controlled mechanical environment. In this review, we highlight the importance of bioreactors as a powerful tool for VF tissue engineering with a focus on the current state of the art of bioreactors designed to mimic phonation in vitro. We discuss the influence of the phonatory environment on the development, function, injury, and healing of the VF tissue and its importance for the development of efficient therapeutic strategies. A concise and comprehensive overview of bioreactor designs, principles, operating parameters, and scalability are presented. An in-depth analysis of VF bioreactor data to date reveals that mechanical stimulation significantly influences cell viability and the expression of proinflammatory and profibrotic genes in vitro. Although the precision and accuracy of bioreactors contribute to generating reliable results, diverse gene expression profiles across the literature suggest that future efforts should focus on the standardization of bioreactor parameters to enable direct comparisons between studies. Impact statement We present a comprehensive review of bioreactors for vocal fold (VF) tissue engineering with a focus on the influence of the phonatory environment on the development, function, injury, and healing of the VFs and the importance of mimicking phonation on engineered VF tissues in vitro. Furthermore, we put forward a strong argument for the continued development of bioreactors in this area with an emphasis on the standardization of bioreactor designs, principles, operating parameters, and oscillatory regimes to enable comparisons between studies.
Collapse
Affiliation(s)
- Ana M. Gracioso Martins
- Joint Department of Biomedical Engineering, College of Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Andreea Biehl
- Joint Department of Biomedical Engineering, College of Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Daphne Sze
- Joint Department of Biomedical Engineering, College of Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Donald O. Freytes
- Joint Department of Biomedical Engineering, College of Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
6
|
Yuan H, Jiao L, Yu N, Duan H, Yu Y, Bai Y. Histone Deacetylase 3-Mediated Inhibition of microRNA-19a-3p Facilitates the Development of Rheumatoid Arthritis-Associated Interstitial Lung Disease. Front Physiol 2020; 11:549656. [PMID: 33343379 PMCID: PMC7746846 DOI: 10.3389/fphys.2020.549656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/02/2020] [Indexed: 11/22/2022] Open
Abstract
Histone deacetylase (HDAC) has been implicated in rheumatoid arthritis (RA) progression. We investigated the roles of histone deacetylase 3 (HDAC3) involved in RA-associated interstitial lung disease (ILD) fibrosis. Firstly, we measured the expression of HDAC3 and interleukin 17 receptor A (IL17RA) in lung tissue samples from normal controls, idiopathic pulmonary fibrosis (IPF) patients, and RA-ILD patients. Next, chromatin immunoprecipitation (ChIP) and dual luciferase reporter assay were employed to detect the interaction between HDAC3 and microRNA-19a-3p (miR-19a-3p) and between miR-19a-3p and IL17RA. Further, immunohistochemistry was used to localize HDAC3 and IL17RA expression in lung tissues. Additionally, functional assays were conducted followed by expression determination of HDAC3, miR-19a-3p, and IL17RA with reverse transcription quantitative PCR (RT-qPCR) and Western blot analysis. The effect of HDAC3 on RA-ILD in the constructed RA-ILD mouse model was also studied based on arthritis assessment. We found overexpressed HDAC3 and IL17RA as well as silenced miR-19a-3p in RA-ILD mouse model and RA-ILD patients. In the mouse model, HDAC3 downregulated miR-19a-3p in lung fibroblasts to promote the progression of RA-ILD fibrosis. In lung fibroblasts of RA-ILD mice, IL17RA was a target gene of miR-19a-3p. miR-19a-3p negatively regulated IL17RA, thereby increasing the expression of fibrosis markers, COL1A1, COL3A1, and FN, in lung fibroblasts of mice. Taken together, HDAC3 upregulated IL17RA expression by targeting miR-19a-3p to facilitate the RA-ILD fibrosis development, which sheds light on a new HDAC3/miR-19a-3p/IL17RA axis functioning in RA-ILD fibrosis.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Rheumatic Nephropathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Li Jiao
- Yanching Institute of Technology, Langfang, China
| | - Nan Yu
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Haifeng Duan
- Department of Imaging, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yong Yu
- Department of Imaging, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yanrong Bai
- Department of Rheumatic Nephropathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
7
|
Abyaneh HS, Regenold M, McKee TD, Allen C, Gauthier MA. Towards extracellular matrix normalization for improved treatment of solid tumors. Theranostics 2020; 10:1960-1980. [PMID: 32042347 PMCID: PMC6993244 DOI: 10.7150/thno.39995] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/09/2019] [Indexed: 12/18/2022] Open
Abstract
It is currently challenging to eradicate cancer. In the case of solid tumors, the dense and aberrant extracellular matrix (ECM) is a major contributor to the heterogeneous distribution of small molecule drugs and nano-formulations, which makes certain areas of the tumor difficult to treat. As such, much research is devoted to characterizing this matrix and devising strategies to modify its properties as a means to facilitate the improved penetration of drugs and their nano-formulations. This contribution presents the current state of knowledge on the composition of normal ECM and changes to ECM that occur during the pathological progression of cancer. It also includes discussion of strategies designed to modify the composition/properties of the ECM as a means to enhance the penetration and transport of drugs and nano-formulations within solid tumors. Moreover, a discussion of approaches to image the ECM, as well as ways to monitor changes in the ECM as a function of time are presented, as these are important for the implementation of ECM-modifying strategies within therapeutic interventions. Overall, considering the complexity of the ECM, its variability within different tissues, and the multiple pathways by which homeostasis is maintained (both in normal and malignant tissues), the available literature - while promising - suggests that improved monitoring of ECM remodeling in vivo is needed to harness the described strategies to their full potential, and match them with an appropriate chemotherapy regimen.
Collapse
Affiliation(s)
- Hoda Soleymani Abyaneh
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, 1650 boul. Lionel-Boulet, Varennes, J3X 1S2, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Maximilian Regenold
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Trevor D. McKee
- STTARR Innovation Centre, University Health Network, 101 College Street Room 7-504, Toronto, Ontario M5G 1L7, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Marc A. Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, 1650 boul. Lionel-Boulet, Varennes, J3X 1S2, Canada
| |
Collapse
|
8
|
Mishriki S, Aithal S, Gupta T, Sahu RP, Geng F, Puri IK. Fibroblasts Accelerate Formation and Improve Reproducibility of 3D Cellular Structures Printed with Magnetic Assistance. RESEARCH (WASHINGTON, D.C.) 2020; 2020:3970530. [PMID: 32776011 PMCID: PMC7395227 DOI: 10.34133/2020/3970530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/18/2020] [Indexed: 12/05/2022]
Abstract
Fibroblasts (mouse, NIH/3T3) are combined with MDA-MB-231 cells to accelerate the formation and improve the reproducibility of 3D cellular structures printed with magnetic assistance. Fibroblasts and MDA-MB-231 cells are cocultured to produce 12.5 : 87.5, 25 : 75, and 50 : 50 total population mixtures. These mixtures are suspended in a cell medium containing a paramagnetic salt, Gd-DTPA, which increases the magnetic susceptibility of the medium with respect to the cells. A 3D monotypic MDA-MB-231 cellular structure is printed within 24 hours with magnetic assistance, whereas it takes 48 hours to form a similar structure through gravitational settling alone. The maximum projected areas and circularities, and cellular ATP levels of the printed structures are measured for 336 hours. Increasing the relative amounts of the fibroblasts mixed with the MDA-MB-231 cells decreases the time taken to form the structures and improves their reproducibility. Structures produced through gravitational settling have larger maximum projected areas and cellular ATP, but are deemed less reproducible. The distribution of individual cell lines in the cocultured 3D cellular structures shows that printing with magnetic assistance yields 3D cellular structures that resemble in vivo tumors more closely than those formed through gravitational settling. The results validate our hypothesis that (1) fibroblasts act as a "glue" that supports the formation of 3D cellular structures, and (2) the structures are produced more rapidly and with greater reproducibility with magnetically assisted printing than through gravitational settling alone. Printing of 3D cellular structures with magnetic assistance has applications relevant to drug discovery, lab-on-chip devices, and tissue engineering.
Collapse
Affiliation(s)
- Sarah Mishriki
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Srivatsa Aithal
- Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Tamaghna Gupta
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Rakesh P. Sahu
- Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
- Department of Materials Science and Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Fei Geng
- Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
- Walter Booth School of Engineering Practice and Technology, Hamilton, Ontario, Canada
| | - Ishwar K. Puri
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
- Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
- Department of Materials Science and Engineering, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
9
|
Cheng YC, Tsai RY, Sung YT, Chen IJ, Tu TY, Mao YY, Wong CS. Melatonin regulation of transcription in the reversal of morphine tolerance: Microarray analysis of differential gene expression. Int J Mol Med 2018; 43:791-806. [PMID: 30569162 PMCID: PMC6317689 DOI: 10.3892/ijmm.2018.4030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
Tolerance and associated hyperalgesia induced by long‑term morphine administration substantially restrict the clinical use of morphine in pain treatment. Melatonin, a neurohormone released by the pineal gland, has been demonstrated to attenuate anti‑nociceptive morphine tolerance. The present study investigates differentially expressed genes in the process of morphine tolerance and altered gene expression subsequent to melatonin treatment in chronic morphine‑infused ratspinal cords. Morphine tolerance was induced in male Wistar rats by intrathecal morphine infusion (the MO group). Melatonin (the MOMa group) was administered to overcome the effects derived by morphine. The mRNA collected from L5‑S3 of the spinal cord was extracted and analysed by rat expression microarray. Principal component analysis and clustering analysis revealed that the overall gene profiles were different in morphine and melatonin treatments. Subsequent to Gene Ontology analysis, the biological processes of differentially expressed genes of MO and MOMa compared with the control group were constructed. Furthermore, a panel of genes exclusively expressed following melatonin treatment and another panel of genes with inverse expression between the MO and MOMa group were also established. Subsequent to PANTHER pathway analysis, a group of genes with inverse expression following melatonin administrated compared with morphine alone were identified. The expression levels of genes of interest were also confirmed using a reverse transcription‑quantitative polymerase chain reaction. The gene panel that was constructed suggests a potential signaling pathway in morphine tolerance development and is valuable for investigating the mechanism of morphine tolerance and the regulatory gene profiles of melatonin treatment. These results may contribute to the discovery of potential drug targets in morphine tolerance treatments in the future.
Collapse
Affiliation(s)
- Yu-Che Cheng
- Proteomics Laboratory, Department of Medical Research, Cathay General Hospital, Taipei 10630, Taiwan, R.O.C
| | - Ru-Yin Tsai
- College of Nursing and Health Sciences, Da‑Yeh University, Changhua 51591, Taiwan, R.O.C
| | - Yen-Tseng Sung
- Proteomics Laboratory, Department of Medical Research, Cathay General Hospital, Taipei 10630, Taiwan, R.O.C
| | - Ing-Jung Chen
- Department of Anesthesiology, Cathay General Hospital, Taipei 10630, Taiwan, R.O.C
| | - Tzu-Yi Tu
- Proteomics Laboratory, Department of Medical Research, Cathay General Hospital, Taipei 10630, Taiwan, R.O.C
| | - Ya-Yuan Mao
- Proteomics Laboratory, Department of Medical Research, Cathay General Hospital, Taipei 10630, Taiwan, R.O.C
| | - Chih-Shung Wong
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| |
Collapse
|
10
|
Ferguson GB, Van Handel B, Bay M, Fiziev P, Org T, Lee S, Shkhyan R, Banks NW, Scheinberg M, Wu L, Saitta B, Elphingstone J, Larson AN, Riester SM, Pyle AD, Bernthal NM, Mikkola HK, Ernst J, van Wijnen AJ, Bonaguidi M, Evseenko D. Mapping molecular landmarks of human skeletal ontogeny and pluripotent stem cell-derived articular chondrocytes. Nat Commun 2018; 9:3634. [PMID: 30194383 PMCID: PMC6128860 DOI: 10.1038/s41467-018-05573-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 07/04/2018] [Indexed: 11/09/2022] Open
Abstract
Tissue-specific gene expression defines cellular identity and function, but knowledge of early human development is limited, hampering application of cell-based therapies. Here we profiled 5 distinct cell types at a single fetal stage, as well as chondrocytes at 4 stages in vivo and 2 stages during in vitro differentiation. Network analysis delineated five tissue-specific gene modules; these modules and chromatin state analysis defined broad similarities in gene expression during cartilage specification and maturation in vitro and in vivo, including early expression and progressive silencing of muscle- and bone-specific genes. Finally, ontogenetic analysis of freshly isolated and pluripotent stem cell-derived articular chondrocytes identified that integrin alpha 4 defines 2 subsets of functionally and molecularly distinct chondrocytes characterized by their gene expression, osteochondral potential in vitro and proliferative signature in vivo. These analyses provide new insight into human musculoskeletal development and provide an essential comparative resource for disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- Gabriel B Ferguson
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA
| | - Ben Van Handel
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA
| | - Maxwell Bay
- Department of Stem Cell Research and Regenerative Medicine, USC, Los Angeles, CA, 90033, USA
| | - Petko Fiziev
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, 90095, USA.,Department of Biological Chemistry, UCLA, Los Angeles, CA, 90095, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, Los Angeles, CA, 90095, USA
| | - Tonis Org
- Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, CA, 90095, USA.,Institute of Molecular and Cell Biology, University of Tartu, Tartu, 51010, Estonia
| | - Siyoung Lee
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA
| | - Ruzanna Shkhyan
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA
| | - Nicholas W Banks
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA
| | - Mila Scheinberg
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA
| | - Ling Wu
- InVitro Cell Research, LLC, Cockeysville, MD, 21030, USA
| | - Biagio Saitta
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA
| | - Joseph Elphingstone
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA
| | - A Noelle Larson
- Departments of Orthopedic Surgery & Biochemistry and Molecular Biology, Center of Regenerative Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Scott M Riester
- Departments of Orthopedic Surgery & Biochemistry and Molecular Biology, Center of Regenerative Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - April D Pyle
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, Los Angeles, CA, 90095, USA
| | - Nicholas M Bernthal
- Department of Orthopaedic Surgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Hanna Ka Mikkola
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, Los Angeles, CA, 90095, USA.,Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, CA, 90095, USA
| | - Jason Ernst
- Department of Biological Chemistry, UCLA, Los Angeles, CA, 90095, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, Los Angeles, CA, 90095, USA.,Computer Science Department, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA.,Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Andre J van Wijnen
- Departments of Orthopedic Surgery & Biochemistry and Molecular Biology, Center of Regenerative Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Michael Bonaguidi
- Department of Stem Cell Research and Regenerative Medicine, USC, Los Angeles, CA, 90033, USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA. .,Department of Stem Cell Research and Regenerative Medicine, USC, Los Angeles, CA, 90033, USA. .,Department of Orthopaedic Surgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
11
|
Sex Differences in Correlation with Gene Expression Levels between Ifi200 Family Genes and Four Sets of Immune Disease-Relevant Genes. J Immunol Res 2018; 2018:1290814. [PMID: 30246031 PMCID: PMC6136564 DOI: 10.1155/2018/1290814] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 01/04/2023] Open
Abstract
Background The HIN-200 family genes in humans have been linked to several autoimmune diseases—particularly to systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Recently, its human counterpart gene cluster, the Ifi200 family in mice, has been linked to spontaneous arthritis disease (SAD). However, many immune-mediated diseases (including RA and SLE) show gender difference. Understanding whether or not and how these genes play a role in sex difference in immune-mediated diseases is essential for diagnosis/treatment. Methods This study takes advantage of the whole genome gene expression profiles of recombinant inbred (RI) strain populations from female and male mice to analyze potential sex differences in a variety of genes in disease pathways. Expression levels and regulatory QTL of Ifi200 family genes between female and male mice were first examined in a large mouse population, including RI strains derived from C57BL/6J, DBA/2J (BXD), and classic inbred strains. Sex similarities and differences were then analyzed for correlations with gene expression levels between genes in the Ifi200 family and four selected gene sets: known immune Ifi200 pathway-related genes, lupus-relevant genes, osteoarthritis- (OA-) and RA-relevant genes, and sex hormone-related genes. Results The expression level of Ifi202b showed the most sex difference in correlation with known immune-related genes (the P value for Ifi202b is 0.0004). Ifi202b also showed gender difference in correlation with selected sex hormone genes, with a P value of 0.0243. When comparing coexpression levels between Ifi200 genes and lupus-relevant genes, Ifi203 and Ifi205 showed significant sex difference (P values: 0.0303 and 0.002, resp.). Furthermore, several key genes (e.g., Csf1r, Ifnb1, IL-20, IL-22, IL-24, Jhdm1d, Csf1r, Ifnb1, IL-20, IL-22, IL-24, and Tgfb2 that regulate sex differences in immune diseases) were discovered. Conclusions Different genes in the Ifi200 family play different roles in sex difference among dissimilar pathways of these four gene groups.
Collapse
|
12
|
Stueckle TA, Davidson DC, Derk R, Kornberg TG, Battelli L, Friend S, Orandle M, Wagner A, Dinu CZ, Sierros KA, Agarwal S, Gupta RK, Rojanasakul Y, Porter DW, Rojanasakul L. Short-Term Pulmonary Toxicity Assessment of Pre- and Post-incinerated Organomodified Nanoclay in Mice. ACS NANO 2018; 12:2292-2310. [PMID: 29451776 PMCID: PMC6357971 DOI: 10.1021/acsnano.7b07281] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Organomodified nanoclays (ONCs) are increasingly used as filler materials to improve nanocomposite strength, wettability, flammability, and durability. However, pulmonary risks associated with exposure along their chemical lifecycle are unknown. This study's objective was to compare pre- and post-incinerated forms of uncoated and organomodified nanoclays for potential pulmonary inflammation, toxicity, and systemic blood response. Mice were exposed via aspiration to low (30 μg) and high (300 μg) doses of preincinerated uncoated montmorillonite nanoclay (CloisNa), ONC (Clois30B), their respective incinerated forms (I-CloisNa and I-Clois30B), and crystalline silica (CS). Lung and blood tissues were collected at days 1, 7, and 28 to compare toxicity and inflammation indices. Well-dispersed CloisNa caused a robust inflammatory response characterized by neutrophils, macrophages, and particle-laden granulomas. Alternatively, Clois30B, I-Clois30B, and CS high-dose exposures elicited a low grade, persistent inflammatory response. High-dose Clois30B exposure exhibited moderate increases in lung damage markers and a delayed macrophage recruitment cytokine signature peaking at day 7 followed by a fibrotic tissue signature at day 28, similar to CloisNa. I-CloisNa exhibited acute, transient inflammation with quick recovery. Conversely, high-dose I-Clois30B caused a weak initial inflammatory signal but showed comparable pro-inflammatory signaling to CS at day 28. The data demonstrate that ONC pulmonary toxicity and inflammatory potential relies on coating presence and incineration status in that coated and incinerated nanoclay exhibited less inflammation and granuloma formation than pristine montmorillonite. High doses of both pre- and post-incinerated ONC, with different surface morphologies, may harbor potential pulmonary health hazards over long-term occupational exposures.
Collapse
Affiliation(s)
- Todd A. Stueckle
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
| | - Donna C. Davidson
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
| | - Ray Derk
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
| | - Tiffany G. Kornberg
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
- Department of Basic Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Lori Battelli
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
| | - Sherri Friend
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
| | - Marlene Orandle
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
| | - Alixandra Wagner
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Cerasela Zoica Dinu
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Konstantinos A. Sierros
- Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Sushant Agarwal
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Rakesh K. Gupta
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Yon Rojanasakul
- Department of Basic Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Dale W. Porter
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
| | - Liying Rojanasakul
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, United States
| |
Collapse
|
13
|
Heart function and thoracic aorta gene expression profiling studies of ginseng combined with different herbal medicines in eNOS knockout mice. Sci Rep 2017; 7:15431. [PMID: 29133875 PMCID: PMC5684410 DOI: 10.1038/s41598-017-15819-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 11/02/2017] [Indexed: 02/06/2023] Open
Abstract
Ginseng, a popular herbal remedy, is often used in combination with other drugs to achieve the maximum therapeutic response. Shenfu (SFI) and Shenmai injection (SMI) have been widely used to treat cardiovascular disease in China. Our study explored the cardiovascular protection of SFI and SMI in eNOS knockout mice to investigate the differences and similarities of the two ginseng-combinations. Transthoracic echocardiography was performed to evaluate the left ventricular structure and function at baseline and 3, 7, and 14 days after drug administration. Agilent Gene Expression microarrays were used to demonstrate the gene expression profiling of the thoracic aorta. Ingenuity Pathway Analysis was performed to evaluate the mechanism improved by SFI and SMI in eNOS knockout mice. Both SFI and SMI could modulate Gadd45 Signaling from TOP15 canonical pathways. Moreover, SFI showed a better effect in the early treatment stage and improved myocardial function via GATA4, GATA6 and COL3A1. Meanwhile, SMI exerted better protective effects at the chronic stage, which may be related to endothelium protection by VEGFA and ACE. The advantage of multi-target by drug combination in progression of complex diseases should be noticed. The appropriate adjustment of drug combination could lead to a better accurate medical care in clinic.
Collapse
|
14
|
Liu F, Jiao Y, Jiao Y, Garcia-Godoy F, Gu W, Liu Q. Sex difference in EGFR pathways in mouse kidney-potential impact on the immune system. BMC Genet 2016; 17:146. [PMID: 27881077 PMCID: PMC5122204 DOI: 10.1186/s12863-016-0449-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 10/15/2016] [Indexed: 12/23/2022] Open
Abstract
Background Epidermal growth factor receptor (Egfr) has been the target of several drugs for cancers. The potential gender differences in genes in the Egfr axis have been suggested in humans and in animal models. Female and male mice from the same recombinant inbred (RI) strain have the same genomic components except the sex difference. A population of different RI mouse strains allows to conduct precise analysis of molecular pathways and regulation of Egfr between female and male mice. Methods The whole genome expression profiles of 70 genetically diverse RI strains of mice were used to compare three major molecular aspects of Egfr gene: the relative expression levels, gene network and expression quantitative trait loci (eQTL) that regulate the expression of Egfr between female and male mice. Results Our data showed that there is a significant sex difference in the expression levels in kidney. A considerable number of genes in the gene network of Egfr are sex differentially expressed. The expression levels of Egfr in mice are statistical significant different between C57BL/6 J (B6) and DBA/2 J (D2) genotypes in male while no difference in female mice. The eQTLs that regulate the expression levels of Egfr between female and male mice are also different. Furthermore, the differential expression levels of Egfr showed significantly different correlations with two known biological traits between male and female mice. Conclusion Overall there is a substantial sex difference in the Egfr pathways in mice. These data may have significant impact on drug target design, development, formulation, and dosage determinant for women and men in clinical trials. Electronic supplementary material The online version of this article (doi:10.1186/s12863-016-0449-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fengxia Liu
- The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, 050011, China.,Department of Orthopaedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yan Jiao
- Department of Orthopaedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yun Jiao
- Department of Neuroscience, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Franklin Garcia-Godoy
- Bioscience Research Center, College of Dentistry, University of Tennessee Health Science Center, 875 Union Avenue, Memphis, TN, USA
| | - Weikuan Gu
- Department of Orthopaedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, USA. .,Research Service, Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN, USA. .,, 956 Court Ave, Memphis, TN, 38163, USA.
| | - Qingyi Liu
- The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, 050011, China.
| |
Collapse
|
15
|
Bertoncello I. Properties of Adult Lung Stem and Progenitor Cells. J Cell Physiol 2016; 231:2582-9. [PMID: 27062064 DOI: 10.1002/jcp.25404] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 04/06/2016] [Indexed: 12/13/2022]
Abstract
The last decade has seen significant progress in understanding the organisation of regenerative cells in the adult lung. Cell-lineage tracing and in vitro clonogenic assays have enabled the identification and characterisation of endogenous lung epithelial stem and progenitor cells. Selective lung injury models, and genetically engineered mice have revealed highly conserved gene networks, factors, signalling pathways, and cellular interactions important in maintaining lung homeostasis and regulating lung regeneration and repair following injury. This review describes the current models of lung epithelial stem and progenitor cell organisation in adult mice, and the impediments encountered in translational studies aiming to identify and characterise their human homologs. J. Cell. Physiol. 231: 2582-2589, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ivan Bertoncello
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Victoria, Australia
| |
Collapse
|
16
|
Wang L, Xiao J, Gu W, Chen H. Sex Difference of Egfr Expression and Molecular Pathway in the Liver: Impact on Drug Design and Cancer Treatments? J Cancer 2016; 7:671-80. [PMID: 27076848 PMCID: PMC4829553 DOI: 10.7150/jca.13684] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 01/22/2016] [Indexed: 12/23/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) has been used as the target in drug design for cancer treatment including the liver cancer. Men and women have different levels of EGFR expression during the life. The whole genome expression profiles of livers of recombinant inbred (RI) strains derived from C57BL/6J (B6) X DBA/2J (D2) were used to compare three major molecular aspects of Egfr gene: the relative expression levels, gene network and eQTLs that regulate the expression of Egfr between female and male mice. Our data suggest that there is a significant difference in the expression levels in the liver between female and male mice. Several important genes in the gene network of Egfr are differentially expressed between female and male mice. The regulatory elements for the expression levels of Egfr between female and male mice are also different. In summary, our data reveals an important sex difference in the Egfr pathways in the liver of the mice. These data may have substantial impact on drug development and dosage determinant for women and men in the clinical trials.
Collapse
Affiliation(s)
- Lishi Wang
- 1. Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, Tennessee, 38163, USA
- 2. Department of Basic Medical Research, Inner Mongolia Medical University, Inner Mongolia, 010110, China
| | - Jianqi Xiao
- 1. Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, Tennessee, 38163, USA
- 3. Center of integrative research, The first Hospital of Qiqihaer City, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China
| | - Weikuan Gu
- 1. Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, Tennessee, 38163, USA
- ✉ Corresponding authors: Weikuan Gu, 956 Court Ave, Memphis, TN 38163, USA. Tel: 1-901-448-2259; ; Hong Chen, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China. Tel: 86-0452-2425981;
| | - Hong Chen
- 3. Center of integrative research, The first Hospital of Qiqihaer City, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China
- ✉ Corresponding authors: Weikuan Gu, 956 Court Ave, Memphis, TN 38163, USA. Tel: 1-901-448-2259; ; Hong Chen, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China. Tel: 86-0452-2425981;
| |
Collapse
|