1
|
Pouyan A, Ghorbanlo M, Eslami M, Jahanshahi M, Ziaei E, Salami A, Mokhtari K, Shahpasand K, Farahani N, Meybodi TE, Entezari M, Taheriazam A, Hushmandi K, Hashemi M. Glioblastoma multiforme: insights into pathogenesis, key signaling pathways, and therapeutic strategies. Mol Cancer 2025; 24:58. [PMID: 40011944 PMCID: PMC11863469 DOI: 10.1186/s12943-025-02267-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary brain tumor in adults, characterized by a poor prognosis and significant resistance to existing treatments. Despite progress in therapeutic strategies, the median overall survival remains approximately 15 months. A hallmark of GBM is its intricate molecular profile, driven by disruptions in multiple signaling pathways, including PI3K/AKT/mTOR, Wnt, NF-κB, and TGF-β, critical to tumor growth, invasion, and treatment resistance. This review examines the epidemiology, molecular mechanisms, and therapeutic prospects of targeting these pathways in GBM, highlighting recent insights into pathway interactions and discovering new therapeutic targets to improve patient outcomes.
Collapse
Affiliation(s)
- Ashkan Pouyan
- Department of Neurosurgery, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Masoud Ghorbanlo
- Department of Anesthesiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Eslami
- Department of Neurosurgery, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Jahanshahi
- Department of Neurosurgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Ziaei
- Department of Neurosurgery, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Salami
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Koorosh Shahpasand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Tohid Emami Meybodi
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Functional Neurosurgery Research Center, Shohada Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Epidemiology, University of Tehran, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
2
|
Yu XH, Guo XN, Li K, Li JW, Wang K, Wang D, Liu BC. The Role of Wnt5a in Inflammatory Diseases. Immunology 2025; 174:203-212. [PMID: 39668514 DOI: 10.1111/imm.13882] [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: 06/22/2024] [Revised: 10/08/2024] [Accepted: 11/22/2024] [Indexed: 12/14/2024] Open
Abstract
Wnt5a plays an important role in cell development and maturation and is closely associated with various diseases, such as malignant tumours, metabolic disorders, fibrosis, growth and development. Recent studies have shown that Wnt5a expression and signal transduction are strongly involved in the inflammatory response. This study comprehensively reviewed the latest research progress on the association between Wnt5a and several inflammatory diseases, such as sepsis, asthma, chronic obstructive pulmonary disease, tuberculosis, rheumatoid arthritis, atherosclerosis and psoriasis vulgare. We elucidated the mechanism by which the Wnt5a protein is involved in the pathogenesis of these diseases, providing a basis for the prevention and treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xin-Hua Yu
- Department of Pediatrics, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Xin-Ning Guo
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kui Li
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Jia-Wei Li
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Kaijin Wang
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bi-Cui Liu
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Xu J, Hu M, Liu L, Xu X, Xu L, Song Y. A transcriptomic analysis of dental pulp stem cell senescence in vitro. Biomed Eng Online 2024; 23:102. [PMID: 39425139 PMCID: PMC11488381 DOI: 10.1186/s12938-024-01298-w] [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: 07/23/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND/PURPOSE The use of human dental pulp stem cells (hDPSCs) as autologous stem cells for tissue repair and regenerative techniques is a significant area of global research. The objective of this study was to investigate the effects of long-term in vitro culture on the multidifferentiation potential of hDPSCs and the potential molecular mechanisms involved. MATERIALS AND METHODS The tissue block method was used to extract hDPSCs from orthodontic-minus-extraction patients, which were then expanded and cultured in vitro for 12 generations. Stem cells from passages three, six, nine, and twelve were selected. Flow cytometry was used to detect the expression of stem cell surface markers, and CCK-8 was used to assess cell proliferation. β-Galactosidase staining was employed to detect cellular senescence, Alizarin Red S staining to assess osteogenic potential, and Oil Red O staining to evaluate lipogenic capacity. RNA sequencing (RNA-seq) was conducted to identify differentially expressed genes in DPSCs and investigate their potential mechanisms. RESULTS With increasing passage numbers, pulp stem cells showed an increase in senescence and a decrease in proliferative capacity and osteogenic-lipogenic multidifferentiation potential. The expression of stem cell surface markers CD34 and CD45 was stable, whereas the expression of CD73, CD90, and CD105 decreased with increasing passages. According to the RNA-seq analysis, the differentially expressed genes CFH, WNT16, HSD17B2, IDI1, and COL5A3 may be associated with stem cell senescence. CONCLUSION Increased in vitro expansion induced cellular senescence in pulp stem cells, which resulted in a reduction in their proliferative capacity and osteogenic-lipogenic differentiation potential. The differential expression of genes such as CFH, WNT16, HSD17B2, IDI1, and COL5A3 may represent a potential mechanism for the induction of cellular senescence in pulp stem cells.
Collapse
Affiliation(s)
- Jidong Xu
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Mingchang Hu
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Longfei Liu
- Qingdao Engineering Vocational College, Qingdao, 266000, China
| | - Xuecheng Xu
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Linlin Xu
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Yu Song
- Department of Orthodontics, Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China.
| |
Collapse
|
4
|
An F, Song J, Chang W, Zhang J, Gao P, Wang Y, Xiao Z, Yan C. Research Progress on the Mechanism of the SFRP-Mediated Wnt Signalling Pathway Involved in Bone Metabolism in Osteoporosis. Mol Biotechnol 2024; 66:975-990. [PMID: 38194214 DOI: 10.1007/s12033-023-01018-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024]
Abstract
Osteoporosis (OP) is a metabolic bone disease linked to an elevated fracture risk, primarily stemming from disruptions in bone metabolism. Present clinical treatments for OP merely alleviate symptoms. Hence, there exists a pressing need to identify novel targets for the clinical treatment of OP. Research indicates that the Wnt signalling pathway is modulated by serum-secreted frizzled-related protein 5 (SFRP5), potentially serving as a pivotal regulator in bone metabolism disorders. Moreover, studies confirm elevated SFRP5 expression in OP, with SFRP5 overexpression leading to the downregulation of Wnt and β-catenin proteins in the Wnt signalling pathway, as well as the expression of osteogenesis-related marker molecules such as RUNX2, ALP, and OPN. Conversely, the opposite has been reported when SFRP5 is knocked out, suggesting that SFRP5 may be a key factor involved in the regulation of bone metabolism via the Wnt signalling axis. However, the molecular mechanisms underlying the action of SFRP5-induced OP have yet to be comprehensively elucidated. This review focusses on the molecular structure and function of SFRP5 and the potential molecular mechanisms of the SFRP5-mediated Wnt signalling pathway involved in bone metabolism in OP, providing reasonable evidence for the targeted therapy of SFRP5 for the prevention and treatment of OP.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jiayi Song
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Weirong Chang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Yujie Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Zhipan Xiao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
5
|
Chiu CSC, Yeh LY, Pan SH, Li SH. Transcriptomic Analysis Reveals Intrinsic Abnormalities in Endometrial Polyps. Int J Mol Sci 2024; 25:2557. [PMID: 38473810 DOI: 10.3390/ijms25052557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Endometrial polyps (EPs) are benign overgrowths of the endometrial tissue lining the uterus, often causing abnormal bleeding or infertility. This study analyzed gene expression differences between EPs and adjacent endometrial tissue to elucidate intrinsic abnormalities promoting pathological overgrowth. RNA sequencing of 12 pairs of EPs and the surrounding endometrial tissue from infertile women revealed 322 differentially expressed genes. Protein-protein interaction network analysis revealed significant alterations in specific signaling pathways, notably Wnt signaling and vascular smooth muscle regulation, suggesting these pathways play critical roles in the pathophysiology of EPs. Wnt-related genes DKK1 and DKKL1 were upregulated, while GPC3, GREM1, RSPO3, SFRP5, and WNT10B were downregulated. Relevant genes for vascular smooth muscle contraction were nearly all downregulated in EPs, including ACTA2, ACTG2, KCNMB1, KCNMB2, MYL9, PPP1R12B, and TAGLN. Overall, the results indicate fundamental gene expression changes promote EP formation through unrestrained growth signaling and vascular defects. The intrinsic signaling abnormalities likely contribute to clinical symptoms of abnormal uterine bleeding and infertility common in EP patients. This analysis provides molecular insights into abnormal endometrial overgrowth to guide improved diagnostic and therapeutic approaches for this troublesome women's health condition. Confirmation of expanded cohorts and further investigations into implicated regulatory relationships are warranted.
Collapse
Affiliation(s)
- Christine Shan-Chi Chiu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei 104, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei 251, Taiwan
| | - Ling-Yu Yeh
- Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei 251, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, Beitou District, Taipei 112, Taiwan
| | - Szu-Hua Pan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Sheng-Hsiang Li
- Department of Medical Research, MacKay Memorial Hospital, Tamsui District, New Taipei 251, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, Beitou District, Taipei 112, Taiwan
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 106, Taiwan
| |
Collapse
|
6
|
Iusupova AO, Pakhtusov NN, Slepova OA, Belenkov YN, Privalova EV, Bure IV, Vetchinkina EA, Nemtsova MV. MiRNA-21a, miRNA-145, and miRNA-221 Expression and Their Correlations with WNT Proteins in Patients with Obstructive and Non-Obstructive Coronary Artery Disease. Int J Mol Sci 2023; 24:17613. [PMID: 38139440 PMCID: PMC10744268 DOI: 10.3390/ijms242417613] [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: 11/10/2023] [Revised: 12/08/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
MicroRNAs and the WNT signaling cascade regulate the pathogenetic mechanisms of atherosclerotic coronary artery disease (CAD) development. OBJECTIVE To evaluate the expression of microRNAs (miR-21a, miR-145, and miR-221) and the role of the WNT signaling cascade (WNT1, WNT3a, WNT4, and WNT5a) in obstructive CAD and ischemia with no obstructive coronary arteries (INOCA). METHOD The cross-sectional observational study comprised 94 subjects. The expression of miR-21a, miR-145, miR-221 (RT-PCR) and the protein levels of WNT1, WNT3a, WNT4, WNT5a, LRP6, and SIRT1 (ELISA) were estimated in the plasma of 20 patients with INOCA (66.5 [62.8; 71.2] years; 25% men), 44 patients with obstructive CAD (64.0 [56.5; 71,0] years; 63.6% men), and 30 healthy volunteers without risk factors for cardiovascular diseases (CVD). RESULTS Higher levels of WNT1 (0.189 [0.184; 0.193] ng/mL vs. 0.15 [0.15-0.16] ng/mL, p < 0.001) and WNT3a (0.227 [0.181; 0.252] vs. 0.115 [0.07; 0.16] p < 0.001) were found in plasma samples from patients with obstructive CAD, whereas the INOCA group was characterized by higher concentrations of WNT4 (0.345 [0.278; 0.492] ng/mL vs. 0.203 [0.112; 0.378] ng/mL, p = 0.025) and WNT5a (0.17 [0.16; 0.17] ng/mL vs. 0.01 [0.007; 0.018] ng/mL, p < 0.001). MiR-221 expression level was higher in all CAD groups compared to the control group (p < 0.001), whereas miR-21a was more highly expressed in the control group than in the obstructive (p = 0.012) and INOCA (p = 0.003) groups. Correlation analysis revealed associations of miR-21a expression with WNT1 (r = -0.32; p = 0.028) and SIRT1 (r = 0.399; p = 0.005) protein levels in all CAD groups. A positive correlation between miR-145 expression and the WNT4 protein level was observed in patients with obstructive CAD (r = 0.436; p = 0.016). Based on multivariate regression analysis, a mathematical model was constructed that predicts the type of coronary lesion. WNT3a and LRP6 were the independent predictors of INOCA (p < 0.001 and p = 0.002, respectively). CONCLUSIONS Activation of the canonical cascade of WNT-β-catenin prevailed in patients with obstructive CAD, whereas in the INOCA and control groups, the activity of the non-canonical pathway was higher. It can be assumed that miR-21a has a negative effect on the formation of atherosclerotic CAD. Alternatively, miR-145 could be involved in the development of coronary artery obstruction, presumably through the regulation of the WNT4 protein. A mathematical model with WNT3a and LRP6 as predictors allows for the prediction of the type of coronary artery lesion.
Collapse
Affiliation(s)
- Alfiya Oskarovna Iusupova
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia (O.A.S.); (Y.N.B.)
| | - Nikolay Nikolaevich Pakhtusov
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia (O.A.S.); (Y.N.B.)
| | - Olga Alexandrovna Slepova
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia (O.A.S.); (Y.N.B.)
| | - Yuri Nikitich Belenkov
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia (O.A.S.); (Y.N.B.)
| | - Elena Vitalievna Privalova
- Department of Hospital Therapy No 1, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia (O.A.S.); (Y.N.B.)
| | - Irina Vladimirovna Bure
- Laboratory of Medical Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (I.V.B.); (E.A.V.); (M.V.N.)
- Research Institute of Molecular and Personalized Medicine, Russian Medical Academy of Continuous Professional Education, 125445 Moscow, Russia
| | - Ekaterina Alexandrovna Vetchinkina
- Laboratory of Medical Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (I.V.B.); (E.A.V.); (M.V.N.)
| | - Marina Vyacheslavovna Nemtsova
- Laboratory of Medical Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (I.V.B.); (E.A.V.); (M.V.N.)
- Laboratory of Epigenetics, Research Centre for Medical Genetics, 115522 Moscow, Russia
| |
Collapse
|
7
|
Russell NX, Burra K, Shah RM, Bottasso-Arias N, Mohanakrishnan M, Snowball J, Ediga HH, Madala SK, Sinner D. Wnt signaling regulates ion channel expression to promote smooth muscle and cartilage formation in developing mouse trachea. Am J Physiol Lung Cell Mol Physiol 2023; 325:L788-L802. [PMID: 37873566 PMCID: PMC11068408 DOI: 10.1152/ajplung.00024.2023] [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: 01/17/2023] [Revised: 08/28/2023] [Accepted: 10/15/2023] [Indexed: 10/25/2023] Open
Abstract
Ion channels play critical roles in the physiology and function of the nervous system and contractile tissue; however, their role in noncontractile tissue and embryonic development has yet to be understood. Tracheobronchomalacia (TBM) and complete tracheal rings (CTR) are disorders affecting the muscle and cartilage of the trachea and bronchi, whose etiology remains poorly understood. We demonstrated that trachealis muscle organization and polarity are disrupted after epithelial ablation of Wntless (Wls), a cargo receptor critical for the Wnt signaling pathway, in developing trachea. The phenotype resembles the anomalous trachealis muscle observed after deletion of ion channel encoding genes in developing mouse trachea. We sought to investigate whether and how the deletion of Wls affects ion channels during tracheal development. We hypothesize that Wnt signaling influences the expression of ion channels to promote trachealis muscle cell assembly and patterning. Deleting Wls in developing trachea causes differential regulation of genes mediating actin binding, cytoskeleton organization, and potassium ion channel activity. Wnt signaling regulates the expression of Kcnj13, Kcnd3, Kcnj8, and Abcc9 as demonstrated by in vitro studies and in vivo analysis in Wnt5a and β-catenin-deficient tracheas. Pharmacological inhibition of potassium ion channels and Wnt signaling impaired contractility of developing trachealis smooth muscle and formation of cartilaginous mesenchymal condensation. Thus, in mice, epithelial-induced Wnt/β-catenin signaling mediates trachealis muscle and cartilage development via modulation of ion channel expression, promoting trachealis muscle architecture, contractility, and cartilaginous extracellular matrix. In turn, ion channel activity may influence tracheal morphogenesis underlying TBM and CTR.NEW & NOTEWORTHY Ion channels play critical roles in the physiology and function of the nervous system and contractile tissue; however, their role in noncontractile tissue and embryonic development has yet to be understood. In this study, we focused on the role of ion channels in the differentiation and patterning of the large airways of the developing respiratory tract. We identify a mechanism by which Wnt-beta-catenin signaling controls levels of ion channel-encoding genes to promote tracheal differentiation.
Collapse
Affiliation(s)
- Nicholas X Russell
- Neonatology and Pulmonary Biology Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati Honors Program, Cincinnati, Ohio, United States
| | - Kaulini Burra
- Neonatology and Pulmonary Biology Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Ronak M Shah
- Neonatology and Pulmonary Biology Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati Honors Program, Cincinnati, Ohio, United States
| | - Natalia Bottasso-Arias
- Neonatology and Pulmonary Biology Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Megha Mohanakrishnan
- Neonatology and Pulmonary Biology Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati Honors Program, Cincinnati, Ohio, United States
| | - John Snowball
- Neonatology and Pulmonary Biology Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Harshavardhana H Ediga
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Satish K Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Debora Sinner
- Neonatology and Pulmonary Biology Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, College of Medicine, Cincinnati, Ohio, United States
| |
Collapse
|
8
|
Russell NX, Burra K, Shah R, Bottasso-Arias N, Mohanakrishnan M, Snowball J, Ediga HH, Madala SK, Sinner D. Wnt signaling regulates ion channel expression to promote smooth muscle and cartilage formation in developing mouse trachea. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.10.523309. [PMID: 36711918 PMCID: PMC9882072 DOI: 10.1101/2023.01.10.523309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Ion channels play critical roles in the physiology and function of the nervous system and contractile tissue; however, their role in non-contractile tissue and embryonic development has yet to be understood. Tracheobronchomalacia (TBM) and complete tracheal rings (CTR) are disorders affecting the muscle and cartilage of the trachea and bronchi, whose etiology remains poorly understood. We demonstrated that trachealis muscle organization and polarity are disrupted after epithelial ablation of Wls, a cargo receptor critical for the Wnt signaling pathway, in developing trachea. The phenotype resembles the anomalous trachealis muscle observed after deletion of ion channel encoding genes in developing mouse trachea. We sought to investigate whether and how the deletion of Wls affects ion channels during tracheal development. We hypothesize that Wnt signaling influences the expression of ion channels to promote trachealis muscle cell assembly and patterning. Deleting Wls in developing trachea causes differential regulation of genes mediating actin binding, cytoskeleton organization, and potassium ion channel activity. Wnt signaling regulated expression of Kcnj13, Kcnd3, Kcnj8, and Abcc9 as demonstrated by in vitro studies and in vivo analysis in Wnt5a and β-catenin deficient tracheas. Pharmacological inhibition of potassium ion channels and Wnt signaling impaired contractility of developing trachealis smooth muscle and formation of cartilaginous mesenchymal condensation. Thus, in mice, epithelial-induced Wnt/β-catenin signaling mediates trachealis muscle and cartilage development via modulation of ion channel expression, promoting trachealis muscle architecture, contractility, and cartilaginous extracellular matrix. In turn, ion channel activity may influence tracheal morphogenesis underlying TBM and CTR.
Collapse
Affiliation(s)
- Nicholas X. Russell
- Neonatology and Pulmonary Biology Perinatal Institute. Cincinnati Children’s Hospital Medical Center and University of Cincinnati Honors Program
| | - Kaulini Burra
- Neonatology and Pulmonary Biology Perinatal Institute. Cincinnati Children’s Hospital Medical Center. Current affiliation: Nationwide Children’s Hospital Columbus OH
| | - Ronak Shah
- Neonatology and Pulmonary Biology Perinatal Institute. Cincinnati Children’s Hospital Medical Center and University of Cincinnati Honors Program Current Affiliation: Renaissance School of Medicine at Stony Brook University
| | - Natalia Bottasso-Arias
- Neonatology and Pulmonary Biology Perinatal Institute. Cincinnati Children’s Hospital Medical Center
| | - Megha Mohanakrishnan
- Neonatology and Pulmonary Biology Perinatal Institute. Cincinnati Children’s Hospital Medical Center and University of Cincinnati Honors Program
| | - John Snowball
- Neonatology and Pulmonary Biology Perinatal Institute. Cincinnati Children’s Hospital Medical Center. Current affiliation: P&G Cincinnati, OH
| | - Harshavardhana H. Ediga
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Satish K Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Debora Sinner
- Neonatology and Pulmonary Biology Perinatal Institute. Cincinnati Children’s Hospital Medical Center and University of Cincinnati, College of Medicine
| |
Collapse
|
9
|
Khan MS, Kim HS, Kim R, Yoon SH, Kim SG. Dysregulated Liver Metabolism and Polycystic Ovarian Syndrome. Int J Mol Sci 2023; 24:ijms24087454. [PMID: 37108615 PMCID: PMC10138914 DOI: 10.3390/ijms24087454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/10/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
A significant fraction of couples around the world suffer from polycystic ovarian syndrome (PCOS), a disease defined by the characteristics of enhanced androgen synthesis in ovarian theca cells, hyperandrogenemia, and ovarian dysfunction in women. Most of the clinically observable symptoms and altered blood biomarker levels in the patients indicate metabolic dysregulation and adaptive changes as the key underlying mechanisms. Since the liver is the metabolic hub of the body and is involved in steroid-hormonal detoxification, pathological changes in the liver may contribute to female endocrine disruption, potentially through the liver-to-ovary axis. Of particular interest are hyperglycemic challenges and the consequent changes in liver-secretory protein(s) and insulin sensitivity affecting the maturation of ovarian follicles, potentially leading to female infertility. The purpose of this review is to provide insight into emerging metabolic mechanisms underlying PCOS as the primary culprit, which promote its incidence and aggravation. Additionally, this review aims to summarize medications and new potential therapeutic approaches for the disease.
Collapse
Affiliation(s)
- Muhammad Sohaib Khan
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si 10326, Republic of Korea
| | - Hee-Sun Kim
- Department of Obstetrics and Gynecology, Dongguk University Ilsan Medical Center, Goyang-si 10326, Republic of Korea
| | - Ranhee Kim
- Department of Obstetrics and Gynecology, Dongguk University Ilsan Medical Center, Goyang-si 10326, Republic of Korea
| | - Sang Ho Yoon
- Department of Obstetrics and Gynecology, Dongguk University Ilsan Medical Center, Goyang-si 10326, Republic of Korea
- Department of Obstetrics and Gynecology, Dongguk University Medical College, Goyang-si 10326, Republic of Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si 10326, Republic of Korea
| |
Collapse
|
10
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
11
|
Xiong Y, Xu Y, Zhou F, Hu Y, Zhao J, Liu Z, Zhai Q, Qi S, Zhang Z, Chen L. Bio-functional hydrogel with antibacterial and anti-inflammatory dual properties to combat with burn wound infection. Bioeng Transl Med 2023; 8:e10373. [PMID: 36684072 PMCID: PMC9842067 DOI: 10.1002/btm2.10373] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/07/2022] [Accepted: 06/12/2022] [Indexed: 01/25/2023] Open
Abstract
Burn infection delays wound healing and increases the burn patient mortality. Consequently, a new dressing with antibacterial and anti-inflammatory dual properties is urgently required for wound healing. In this study, we propose a combination of methacrylate gelatin (GelMA) hydrogel system with silver nanoparticles embed in γ-cyclodextrin metal-organic frameworks (Ag@MOF) and hyaluronic acid-epigallocatechin gallate (HA-E) for the burn wound infection treatment. Ag@MOF is used as an antibacterial agent and epigallocatechin gallate (EGCG) has exhibited biological properties of anti-inflammation and antibacterial. The GelMA/HA-E/Ag@MOF hydrogel enjoys suitable physical properties and sustained release of Ag+. Meanwhile, the hydrogel has excellent biocompatibility and could promote macrophage polarization from M1 to M2. In vivo wound healing evaluations further demonstrate that the GelMA/HA-E/Ag@MOF hydrogel reduces the number of the bacterium efficiently, accelerates wound healing, promotes early angiogenesis, and regulates immune reaction. A further evaluation indicates that the noncanonical Wnt signal pathway is significantly activated in the GelMA/HA-E/Ag@MOF hydrogel treated group. In conclusion, the GelMA/HA-E/Ag@MOF hydrogel could serve as a promising multifunctional dressing for the burn wound healing.
Collapse
Affiliation(s)
- Yahui Xiong
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Yingbin Xu
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Fei Zhou
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Yanke Hu
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Jingling Zhao
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Zhonghua Liu
- South China Agricultural UniversityGuangzhouChina
| | - Qiyi Zhai
- ZhuJiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shaohai Qi
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Zhaoqiang Zhang
- Department of Oral and Maxillofacial SurgeryStomatological Hospital, Southern Medical UniversityGuangzhouChina
| | - Lei Chen
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| |
Collapse
|
12
|
Wang W, Shang W, Zou J, Liu K, Liu M, Qiu X, Zhang H, Wang K, Wang N. ZNF667 facilitates angiogenesis after myocardial ischemia through transcriptional regulation of VASH1 and Wnt signaling pathway. Int J Mol Med 2022; 50:129. [PMID: 36043524 PMCID: PMC9448299 DOI: 10.3892/ijmm.2022.5185] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/09/2022] [Indexed: 11/24/2022] Open
Abstract
Zinc finger protein 667 (ZNF667, also referred as Mipu1), a widely expressed KRAB/C2H2-type zinc finger transcription factor, can protect against hypoxic-ischemic myocardial injury. Pro-angiogenesis is regarded as a promising strategy for the treatment of acute myocardial infarction (AMI). However, whether ZNF667 is involved in the angiogenesis following AMI remains to be elucidated. The present study reported that the expression of ZNF667 in CD31-positive endothelial cells (ECs) was upregulated in the heart of AMI mice. Hypoxic challenge (1% oxygen) promoted the mRNA and protein expression of ZNF667 in the human umbilical vein endothelial cells (HUVECs) in a time-dependent manner. Moreover, ZNF667 promoted hypoxia-induced invasion and tube formation of HUVECs. Mechanically, ZNF667 could directly bind to the promoter of anti-angiogenic gene VASH1 and inhibit its expression. Consequently, VASH1 overexpression abolished hypoxic challenge or ZNF667 overexpression-induced invasion and tube formation of HUVECs. Further bioinformatic analyses suggested that overexpression of ZNF667 or knockdown of VASH1-induced differentially expressed genes in HUVECs were greatly enriched in the Wnt signaling pathway (DAAM1, LEF1, RAC2, FRAT1, NFATc2 and WNT5A). Together, these data suggested that ZNF667 facilitates myocardial ischemia-driven angiogenesis through transcriptional repression of VASH1 and regulation of Wnt signaling pathway.
Collapse
Affiliation(s)
- Wenmei Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Weite Shang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jiang Zou
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Meidong Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaoqin Qiu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
13
|
Wang C, Qu K, Wang J, Qin R, Li B, Qiu J, Wang G. Biomechanical regulation of planar cell polarity in endothelial cells. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166495. [PMID: 35850177 DOI: 10.1016/j.bbadis.2022.166495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/03/2023]
Abstract
Cell polarity refers to the uneven distribution of certain cytoplasmic components in a cell with a spatial order. The planar cell polarity (PCP), the cell aligns perpendicular to the polar plane, in endothelial cells (ECs) has become a research hot spot. The planar polarity of ECs has a positive significance on the regulation of cardiovascular dysfunction, pathological angiogenesis, and ischemic stroke. The endothelial polarity is stimulated and regulated by biomechanical force. Mechanical stimuli promote endothelial polarization and make ECs produce PCP to maintain the normal physiological and biochemical functions. Here, we overview recent advances in understanding the interplay and mechanism between PCP and ECs function involved in mechanical forces, with a focus on PCP signaling pathways and organelles in regulating the polarity of ECs. And then showed the related diseases caused by ECs polarity dysfunction. This study provides new ideas and therapeutic targets for the treatment of endothelial PCP-related diseases.
Collapse
Affiliation(s)
- Caihong Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Jing Wang
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Rui Qin
- College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Bingyi Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| |
Collapse
|
14
|
Geng B, Zhu Y, Yuan Y, Bai J, Dou Z, Sui A, Luo W. Artesunate Suppresses Choroidal Melanoma Vasculogenic Mimicry Formation and Angiogenesis via the Wnt/CaMKII Signaling Axis. Front Oncol 2021; 11:714646. [PMID: 34476217 PMCID: PMC8406848 DOI: 10.3389/fonc.2021.714646] [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: 05/25/2021] [Accepted: 07/26/2021] [Indexed: 01/14/2023] Open
Abstract
Angiogenesis and vasculogenic mimicry (VM) are considered to be the main processes to ensure tumor blood supply during the proliferation and metastasis of choroidal melanoma (CM). The traditional antimalarial drug artesunate (ART) has some potential anti-CM effects; however, the underlying mechanisms remain unclarified. Recent studies have shown that the Wnt5a/calmodulin-dependent kinase II (CaMKII) signaling pathway has a close correlation with angiogenesis and VM formation. This study demonstrated that ART eliminated VM formation by inhibiting the aforementioned signaling pathway in CM cells. The microvessel sprouting of the mouse aortic rings and the microvessel density of chicken chorioallantoic membrane (CAM) decreased significantly after ART treatment. VM formation assay and periodic acid schiff (PAS) staining revealed that ART inhibited VM formation in CM. Moreover, ART downregulated the expression levels of the angiogenesis-related proteins vascular endothelial growth factor receptor (VEGFR) 2, platelet-derived growth factor receptor (PDGFR) and vascular endothelial growth factor (VEGF) A, and VM-related proteins ephrin type-A receptor (EphA) 2 and vascular endothelial (VE)-cadherin. The expression of hypoxia-inducible factor (HIF)-1α, Wnt5a, and phosphorylated CaMKII was also downregulated after ART treatment. In addition, we further demonstrated that ART inhibited the proliferation, migration, and invasion of OCM-1 and C918 cells. Collectively, our results suggested that ART inhibited angiogenesis and VM formation of choroidal melanoma likely by regulating the Wnt5a/CaMKII signaling pathway. These findings further supported the feasibility of ART for cancer therapy.
Collapse
Affiliation(s)
- Bochao Geng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuanzhang Zhu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingying Yuan
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jingyi Bai
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhizhi Dou
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aihua Sui
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenjuan Luo
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
15
|
Soliman AM, Das S, Mahakkanukrauh P. Inflammatory Molecular Mediators and Pathways Involved in Vascular Aging and Stroke: A Comprehensive Review. Curr Med Chem 2021; 29:5522-5542. [PMID: 34488579 DOI: 10.2174/0929867328666210901122359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/01/2021] [Accepted: 07/23/2021] [Indexed: 11/22/2022]
Abstract
There is an increase in the incidence of cardiovascular diseases with aging and it is one of the leading causes of death worldwide. The main cardiovascular pathologies include atherosclerosis, stroke, myocardial infarction, hypertension and stroke. Chronic inflammation is one of the significant contributors to the age-related vascular diseases. Therefore, it is important to understand the molecular mechanisms of the persistent inflammatory conditions occurring in the blood vessels as well as the signaling pathways involved. Herein, we performed an extant search of literature involving PubMed, ISI, WoS and Scopus databases for retrieving all relevant articles with the most recent findings illustrating the potential role of various inflammatory mediators along with their proposed activated pathways in the pathogenesis and progression of vascular aging. We also highlight the major pathways contributing to age-related vascular disorders. The outlined molecular mechanisms, pathways and mediators of vascular aging represent potential drug targets that can be utilized to inhibit and/or slow the pathogenesis and progression of vascular aging.
Collapse
Affiliation(s)
- Amro M Soliman
- Department of Biological Sciences-Physiology, Cell and Developmental Biology, University of Alberta, Edmonton, AB T6G 2R3. Canada
| | - Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, P.C. 123, Al Khoud, Muscat. Oman
| | - Pasuk Mahakkanukrauh
- Department of Anatomy & Excellence center of Osteology Research and Training, Cadaveric Surgical and Training Center, Chiang Mai University, Chiang Mai 50200. Thailand
| |
Collapse
|
16
|
Spicer LJ. Wingless-type mouse mammary tumor virus integration site regulation of bovine theca cells. J Anim Sci 2021; 99:6309027. [PMID: 34166505 DOI: 10.1093/jas/skab197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/22/2021] [Indexed: 11/14/2022] Open
Abstract
Ovarian paracrine mediation by components of the wingless-type mouse mammary tumor virus integration site ligands (WNT1 to 11) and their receptors, frizzled family members (FZD1 to 10), has been proposed. Secreted truncated forms of FZD proteins (e.g., secreted frizzled-related protein 4 [SFRP4]) block the action of WNT ligands. Dickkopf-1 (DKK1) is another WNT antagonist, and R-spondin-1 (RSPO1) is one of a group of four secreted proteins that enhance WNT/β-catenin signaling. Our hypothesis was that granulosa cells signal theca cells (TCs) via SFRP4, DKK1, RSPO1, and WNT secretion to regulate TC differentiation and proliferation. Therefore, in vitro experiments were conducted to study the effects of WNT family member 3A (WNT3A), WNT5A, RSPO1, DKK1, insulin-like growth factor 1 (IGF1), bone morphogenetic protein 7 (BMP7), Indian hedgehog (IHH), and fibroblast growth factor 9 (FGF9) on bovine TC proliferation and steroidogenesis. TCs of large (8 to 20 mm) and small (3 to 6 mm) follicles were collected from bovine ovaries; TC monolayers were established in vitro and treated with various doses of recombinant human WNT3A, WNT5A, RSPO1, DKK1, IGF1, FGF9, BMP7, IHH, and/or ovine luteinizing hormone (LH) in serum-free medium for 48 h. In experiment 1, using LH-treated TC, IGF1, IHH, and WNT3A increased (P < 0.05) cell numbers and androstenedione production, whereas WNT3A and BMP7 inhibited (P < 0.05) progesterone production. In experiment 2, FGF9 blocked (P < 0.05) the WNT3A-induced increase in androstenedione production in LH plus IGF1-treated TC. In experiment 3, RSPO1 further increased (P < 0.05) LH plus IGF1-induced progesterone and androstenedione production. In experiment 4, SFRP4 and DKK1 alone had no significant effect on TC proliferation or progesterone production of large-follicle TC but both blocked the inhibitory effect of WNT5A on androstenedione production. In contrast, DKK1 alone inhibited (P < 0.05) small-follicle TC androstenedione production whereas SFRP4 was without effect. We conclude that the ovarian TC WNT system is functional in cattle, with WNT3A increasing proliferation and androstenedione production of TC.
Collapse
Affiliation(s)
- Leon J Spicer
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
17
|
Dual role of WNT5A in promoting endothelial differentiation of glioma stem cells and angiogenesis of glioma derived endothelial cells. Oncogene 2021; 40:5081-5094. [PMID: 34188250 DOI: 10.1038/s41388-021-01922-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 01/11/2023]
Abstract
Glioma is a devastating cancer with a rich vascular network. No anti-angiogenic treatment is available for prolonging the overall survival of glioma patients. Recent studies have demonstrated that the endothelial differentiation of glioma stem cells (GSCs) into glioma-derived endothelial cells (GDECs) may be a novel target for anti-angiogenic therapy in glioma; however, the underlying mechanisms of this process remain unknown. Here, we report that wingless-related integration site (WNT) family member 5A (WNT5A) plays significant roles in GSC endothelial differentiation and GDECs angiogenesis. WNT5A is preferentially secreted by GDECs, and inhibition of WNT5A suppresses angiogenesis and tumorigenesis in GDECs. Silencing of WNT5A in GDECs also disrupts the impact of GDECs on stimulating GSC endothelial differentiation. Frizzled-4 is a receptor that mediates the effect of WNT5A on GSC endothelial differentiation and angiogenesis of GDECs via GSK3β/β-catenin/epithelial-mesenchymal transition signalling. The shWNT5A@cRGD-DDD liposomes, targeting WNT5A, exert anti-angiogenic effects in vivo. In this study, we identified that WNT5A has a dual functional role in modulating the endothelial differentiation of GSCs and angiogenesis of GDECs, indicating that WNT5A is a potential target for anti-angiogenesis-based therapeutics in glioma.
Collapse
|
18
|
Expression pattern of WNT5A among Egyptian patients with psoriasis treated with platelet-rich plasma versus conventional therapy: Toward a better understanding of the disease. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Zhao Y, Hu J, Sun X, Yang K, Yang L, Kong L, Zhang B, Li F, Li C, Shi B, Hu K, Sun A, Ge J. Loss of m6A demethylase ALKBH5 promotes post-ischemic angiogenesis via post-transcriptional stabilization of WNT5A. Clin Transl Med 2021; 11:e402. [PMID: 34047466 PMCID: PMC8087997 DOI: 10.1002/ctm2.402] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/11/2021] [Accepted: 04/18/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Post-ischemic angiogenesis is critical for blood flow recovery and ischemic tissue repair. N6-methyladenosine (m6A) plays essential roles in numerous biological processes. However, the impact and connected mechanism of m6A on post-ischemic angiogenesis are not fully understood. METHODS AlkB homolog 5 (ALKBH5) was screened out among several methyltransferases and demethylases involved in dynamic m6A regulation. Cardiac microvascular endothelial cells (CMECs) angiogenesis and WNT family member 5A (WNT5A) stability were analyzed upon ALKBH5 overexpression with adenovirus or knockdown with small interfering RNAs in vitro. The blood flow recovery, capillary, and small artery densities were evaluated in adeno-associated virus (AAV)-ALKBH5 overexpression or ALKBH5 knockout (KO) mice in a hind-limb ischemia model. The same experiments were conducted to explore the translational value of transient silencing of ALKBH5 with adenovirus. RESULTS ALKBH5 was significantly upregulated in hypoxic CMECs and led to a global decrease of m6A level. ALKBH5 overexpression further reduced m6A level in normoxic and hypoxic CMECs, impaired proliferation, migration, and tube formation only in hypoxic CMECs. Conversely, ALKBH5 knockdown preserved m6A levels and promoted angiogenic phenotypes in hypoxic but not in normoxic CMECs. Mechanistically, ALKBH5 regulated WNT5A expression through post-transcriptional mRNA modulation in an m6A-dependent manner, which decreased its stability and subsequently impeded angiogenesis in hypoxic CMECs. Furthermore, ALKBH5 overexpression hindered blood flow recovery and reduced CD31 and alpha-smooth muscle actin expression in hind-limb ischemia mice. As expected, ALKBH5-KO mice exhibited improved blood flow recovery, increased capillary, and small artery densities after hind-limb ischemia, and similar beneficial effects were observed in mice with transient adenoviral ALKBH5 gene silencing. CONCLUSION We demonstrate that ALKBH5 is a negative regulator of post-ischemic angiogenesis via post-transcriptional modulation and destabilization of WNT5A mRNA in an m6A-dependent manner. Targeting ALKBH5 may be a potential therapeutic option for ischemic diseases, including peripheral artery disease.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
- NHC Key Laboratory of Viral Heart DiseasesShanghaiChina
- Key Laboratory of Viral Heart DiseasesChinese Academy of Medical SciencesShanghaiChina
- Department of CardiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Jingjing Hu
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
- NHC Key Laboratory of Viral Heart DiseasesShanghaiChina
- Key Laboratory of Viral Heart DiseasesChinese Academy of Medical SciencesShanghaiChina
| | - Xiaolei Sun
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
- NHC Key Laboratory of Viral Heart DiseasesShanghaiChina
- Key Laboratory of Viral Heart DiseasesChinese Academy of Medical SciencesShanghaiChina
| | - Kun Yang
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
- NHC Key Laboratory of Viral Heart DiseasesShanghaiChina
- Key Laboratory of Viral Heart DiseasesChinese Academy of Medical SciencesShanghaiChina
| | - Lebing Yang
- Department of CardiologyWenzhou Medicial UniversityWenzhouChina
| | - Lingqiu Kong
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
- NHC Key Laboratory of Viral Heart DiseasesShanghaiChina
- Key Laboratory of Viral Heart DiseasesChinese Academy of Medical SciencesShanghaiChina
| | - Beijian Zhang
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
- NHC Key Laboratory of Viral Heart DiseasesShanghaiChina
- Key Laboratory of Viral Heart DiseasesChinese Academy of Medical SciencesShanghaiChina
| | - Fuhai Li
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
| | - Chaofu Li
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
| | - Bei Shi
- Department of CardiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Kai Hu
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
| | - Aijun Sun
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
- NHC Key Laboratory of Viral Heart DiseasesShanghaiChina
- Key Laboratory of Viral Heart DiseasesChinese Academy of Medical SciencesShanghaiChina
| | - Junbo Ge
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
- NHC Key Laboratory of Viral Heart DiseasesShanghaiChina
- Key Laboratory of Viral Heart DiseasesChinese Academy of Medical SciencesShanghaiChina
- Department of CardiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
20
|
Sun Q, Liu S, Feng J, Kang Y, Zhou Y, Guo S. Current Status of MicroRNAs that Target the Wnt Signaling Pathway in Regulation of Osteogenesis and Bone Metabolism: A Review. Med Sci Monit 2021; 27:e929510. [PMID: 33828067 PMCID: PMC8043416 DOI: 10.12659/msm.929510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The directional differentiation of bone mesenchymal stem cells (BMSCs) is regulated by a variety of transcription factors and intracellular signaling pathways. In the past, it was thought that the directional differentiation of BMSCs was related to transforming growth factors, such as bone morphogenetic protein (BMP) and MAPK pathway. However, in recent years, some scholars have pointed out that the Wnt signaling pathway, which is a necessary complex network of protein interactions for biological growth and development, takes a significant role in this process and plays a major part in regulating the development of osteoblasts by exerting signal transduction into cells. Also, they have proved the Wnt protein therapeutic truly have positive effects on the viability and osteogenic capacity of bone graft. Recent studies have shown that microRNAs (miRNAs) play an important regulatory role in this process. MiRNAs such as miRNA-218, miRNA-335, miRNA-29, microRNA-30 and other miRNAs exert negative or positive effects on some crucial molecules in the Wnt/β-catenin pathway, which in turn affect bone metabolism and osteopathy. Thus, miRNAs have been suggested as therapeutic targets for some metabolic bone diseases. This article aims to provide an update on the current status of microRNAs that target the Wnt signaling pathway in the regulation of osteogenesis and bone metabolism and includes a discussion of future areas of research, which can be a theoretical basis for bone metabolism-related diseases.
Collapse
Affiliation(s)
- Qiang Sun
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Siyu Liu
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Jingyi Feng
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yue Kang
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - You Zhou
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Shu Guo
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
21
|
Zhang F, Sun P, Yuan N. miR-141-3p Reduces Cell Migration and Proliferation in an In Vitro Modelof Atherosclerosis by Targeting Wnt5a. J INVEST SURG 2021; 35:598-604. [PMID: 33818249 DOI: 10.1080/08941939.2021.1904467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Atherosclerosis (AS) is a type of chronic vascular disease that is also a leading cause of numerous cardiovascular diseases in humans. The biomolecules responsible for the roles of microRNA (miR)-141-3p during AS development are less understood. METHODS The relation between Wnt5a and miR-141-3p was predicted using bioinformatics software TargetScan 7.1, and confirmed via dual luciferase reporter assay. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and immunoblotting were conducted for examining miR-141-3p and Wingless and Int-1 (Wnt)5a expression levels. Additionally, transwell migration and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays were conducted for analyzing cell migration and proliferation, respectively. RESULTS miR-141-3p was decreased in oxidized low-density lipoprotein (ox-LDL)-treated human vascular smooth muscle cells (VSMCs). Pretreatment with miR-141-3p mimic inhibited cell migration and proliferation in ox-LDL-induced VSMCs. Wnt5a was verified to act as the target of miR-141-3p in VSMCs. pcDNA3-Wnt5a partially reversed the effects of miR-141-3p mimic in ox-LDL-stimulated VSMCs. CONCLUSION miR-141-3p mimic decreased the damage in an AS model by targeting Wnt5a, thereby presenting a novel potential therapeutic target for treating AS.
Collapse
Affiliation(s)
- Futao Zhang
- Department of Cardiothoracic Vascular Surgery, Qingdao Chengyang People's Hospital, Qingdao, China
| | - Pishan Sun
- Department of Cardiothoracic Vascular Surgery, Qingdao Chengyang People's Hospital, Qingdao, China
| | - Ningning Yuan
- Department of Rehabilitation Medicine, Qingdao Chengyang People's Hospital, Qingdao, China
| |
Collapse
|
22
|
Zhang N, Hu L, Liu J, Yang W, Li Y, Pan J. Wnt Signaling Regulates the Lymphatic Endothelial Transdifferentiation of Adipose-Derived Stromal Cells In Vitro. Cell Reprogram 2021; 23:117-126. [PMID: 33780637 DOI: 10.1089/cell.2020.0058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Lymphedema is a chronic, progressive disease that causes pain as well as heavy economic burdens to patients. Reconstruction of the impaired lymphatic system is the key to treat lymphedema. Currently, there is no cure, but mesenchymal stromal cells show promising potential for lymphatic endothelial regeneration. Adipose-derived stromal cells (ADSCs) have been proved to support lymphangiogenesis both in vivo and in vitro. However, the mechanism in vascular endothelial growth factor C-induced (VEGF-C-induced) lymphatic endothelial transdifferentiation of ADSCs remains unknown. In this study, we show a novel link between the Wingless and int-1 (Wnt) pathway and the lymphatic endothelial differentiation process. We used LiCl to activate Wnt and DKK-1 to inhibit Wnt. Compared with the Wnt inhibition group and the control groups, the Wnt activation group produced more lymphatic endothelial cell (LEC)-related mRNA and proteins. Besides, Wnt-activated ADSCs formed longer tubes in two-dimensional culture and promoted the growth of lymphatic vessels in a three-dimensional transwell ADSC-LEC co-culture system. Our results demonstrated that activation of Wnt during the lymphatic endothelial transdifferentiation of ADSCs would enhance the efficacy of VEGF-C treatment. We anticipate our assay to expand our knowledge of Wnt in cell transdifferentiation and lay a foundation for future efforts to explore a novel and effective ADSC-based therapy for lymphedema.
Collapse
Affiliation(s)
- Nian Zhang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liru Hu
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiyuan Liu
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenbin Yang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ye Li
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Wang J, Gong M, Zuo S, Xu J, Paul C, Li H, Liu M, Wang YG, Ashraf M, Xu M. WNT11-Conditioned Medium Promotes Angiogenesis through the Activation of Non-Canonical WNT-PKC-JNK Signaling Pathway. Genes (Basel) 2020; 11:E1277. [PMID: 33137935 PMCID: PMC7694138 DOI: 10.3390/genes11111277] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/25/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We demonstrated that the transduction of Wnt11 into mesenchymal stem cells (MSCs) (MSCWnt11) promotes these cells differentiation into cardiac phenotypes. In the present study, we investigated the paracrine effects of MSCWnt11 on cardiac function and angiogenesis. METHODS AND RESULTS Conditioned medium was collected from MSCWnt11 (CdMWnt11) and their control cells (CdMGFP). CdMWnt11, especially obtained from MSCWnt11 exposed to hypoxia, significantly promoted human umbilical vein endothelial cells (HUVECs) migration and increased capillary-like tube (CLT) formation, which was blocked by Wnt11 neutralizing antibody. Wnt11 protein was significantly higher in CdMWnt11 compared to that in CdMGFP. Directly treating HUVECs with recombinant Wnt11 protein significantly increased CLT formation, which was abrogated by treating cells with the JNK inhibitor SP600125, as well as the PKC inhibitor Calphostin-C. Moreover, the transfection of Wnt11 to HUVECs (HWnt11) significantly increased CLT formation and HUVEC migration, as well as upregulated p-pan-PKC and p-JNK expression. Injection of CdMWnt11 into the peri-infarct region in a rat acute myocardial infarction (AMI) model significantly improved cardiac function, reduced infarct size, and increased myocardial blood flow and blood vessel density in the ischemic area. CONCLUSION Wnt11 released from MSCWnt11 increased angiogenesis and improved cardiac function via non-canonical Wnt-PKC-JNK dependent pathways.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA; (J.W.); (M.G.); (S.Z.); (J.X.); (C.P.); (H.L.); (M.L.); (Y.-G.W.); (M.A.)
| |
Collapse
|
24
|
Liu Y, Neogi A, Mani A. The role of Wnt signalling in development of coronary artery disease and its risk factors. Open Biol 2020; 10:200128. [PMID: 33081636 PMCID: PMC7653355 DOI: 10.1098/rsob.200128] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/23/2020] [Indexed: 02/05/2023] Open
Abstract
The Wnt signalling pathways are composed of a highly conserved cascade of events that govern cell differentiation, apoptosis and cell orientation. Three major and distinct Wnt signalling pathways have been characterized: the canonical Wnt pathway (or Wnt/β-catenin pathway), the non-canonical planar cell polarity pathway and the non-canonical Wnt/Ca2+ pathway. Altered Wnt signalling pathway has been associated with diverse diseases such as disorders of bone density, different malignancies, cardiac malformations and heart failure. Coronary artery disease is the most common type of heart disease in the United States. Atherosclerosis is a multi-step pathological process, which starts with lipid deposition and endothelial cell dysfunction, triggering inflammatory reactions, followed by recruitment and aggregation of monocytes. Subsequently, monocytes differentiate into tissue-resident macrophages and transform into foam cells by the uptake of modified low-density lipoprotein. Meanwhile, further accumulations of lipids, infiltration and proliferation of vascular smooth muscle cells, and deposition of the extracellular matrix occur under the intima. An atheromatous plaque or hyperplasia of the intima and media is eventually formed, resulting in luminal narrowing and reduced blood flow to the myocardium, leading to chest pain, angina and even myocardial infarction. The Wnt pathway participates in all different stages of this process, from endothelial dysfunction to lipid deposit, and from initial inflammation to plaque formation. Here, we focus on the role of Wnt cascade in pathophysiological mechanisms that take part in coronary artery disease from both clinical and experimental perspectives.
Collapse
Affiliation(s)
- Ya Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Arpita Neogi
- Yale Cardiovascular Genetics Program, Yale University, New Haven, CT, USA
| | - Arya Mani
- Yale Cardiovascular Genetics Program, Yale University, New Haven, CT, USA
- Yale Cardiovascular Research Center, Department of Medicine, Yale University, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
25
|
Kim JA, Choi KM. Newly Discovered Adipokines: Pathophysiological Link Between Obesity and Cardiometabolic Disorders. Front Physiol 2020; 11:568800. [PMID: 32982804 PMCID: PMC7492654 DOI: 10.3389/fphys.2020.568800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/14/2020] [Indexed: 12/21/2022] Open
Abstract
With the increasing prevalence of obesity, obesity-related problems such as cardiometabolic disorders (CMD), are also rapidly increasing. To prevent and alleviate the progressive course of CMD, it is important to discover the pathophysiological mechanisms between obesity and CMD. Adipose tissue is now recognized as an active endocrine organ that releases adipokines. Adipokines play a pivotal role in chronic low-grade inflammation, oxidative stress, and impaired insulin signaling, contributing to metabolic derangement and leading to CMD. Recent studies have provided substantial evidence supporting the association between adipokines and CMD. In this review, we highlight the pathophysiological action of adipokines in CMD that includes metabolic syndrome, type 2 diabetes, non-alcoholic fatty liver disease, and cardiovascular diseases. We focused on translational and clinical research of novel adipokines associated with metabolic and cardiovascular regulation. Exploration of the role of these adipokines connecting obesity and CMD may provide a perspective on adipokine-based therapeutic implications for CMD.
Collapse
Affiliation(s)
- Jung A Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
26
|
Exner EC, Geurts AM, Hoffmann BR, Casati M, Stodola T, Dsouza NR, Zimmermann M, Lombard JH, Greene AS. Interaction between Mas1 and AT1RA contributes to enhancement of skeletal muscle angiogenesis by angiotensin-(1-7) in Dahl salt-sensitive rats. PLoS One 2020; 15:e0232067. [PMID: 32324784 PMCID: PMC7179868 DOI: 10.1371/journal.pone.0232067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
The heptapeptide angiotensin-(1-7) (Ang-(1-7)) is protective in the cardiovascular system through its induction of vasodilator production and angiogenesis. Despite acting antagonistically to the effects of elevated, pathophysiological levels of angiotensin II (AngII), recent evidence has identified convergent and beneficial effects of low levels of both Ang-(1-7) and AngII. Previous work identified the AngII receptor type I (AT1R) as a component of the protein complex formed when Ang-(1-7) binds its receptor, Mas1. Importantly, pharmacological blockade of AT1R did not alter the effects of Ang-(1-7). Here, we use a novel mutation of AT1RA in the Dahl salt-sensitive (SS) rat to test the hypothesis that interaction between Mas1 and AT1R contributes to proangiogenic Ang-(1-7) signaling. In a model of hind limb angiogenesis induced by electrical stimulation, we find that the restoration of skeletal muscle angiogenesis in SS rats by Ang-(1-7) infusion is impaired in AT1RA knockout rats. Enhancement of endothelial cell (EC) tube formation capacity by Ang-(1-7) is similarly blunted in AT1RA mutant ECs. Transcriptional changes elicited by Ang-(1-7) in SS rat ECs are altered in AT1RA mutant ECs, and tandem mass spectrometry-based proteomics demonstrate that the protein complex formed upon binding of Ang-(1-7) to Mas1 is altered in AT1RA mutant ECs. Together, these data support the hypothesis that interaction between AT1R and Mas1 contributes to proangiogenic Ang-(1-7) signaling.
Collapse
MESH Headings
- Angiotensin I/metabolism
- Animals
- Electric Stimulation
- Male
- Mass Spectrometry
- Models, Animal
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/metabolism
- Mutation
- Neovascularization, Physiologic
- Peptide Fragments/metabolism
- Proteomics
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/metabolism
- Rats
- Rats, Inbred Dahl
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Eric C. Exner
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Aron M. Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Brian R. Hoffmann
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Bioengineering, Medical College of Wisconsin and Marquette University, Milwaukee, Wisconsin, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Marc Casati
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Timothy Stodola
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Nikita R. Dsouza
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Michael Zimmermann
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Julian H. Lombard
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Andrew S. Greene
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| |
Collapse
|
27
|
WNT5a-ROR Signaling Is Essential for Alveologenesis. Cells 2020; 9:cells9020384. [PMID: 32046118 PMCID: PMC7072327 DOI: 10.3390/cells9020384] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/14/2022] Open
Abstract
WNT5a is a mainly “non-canonical” WNT ligand whose dysregulation is observed in lung diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and asthma. Germline deletion of Wnt5a disrupts embryonic lung development. However, the temporal-specific function of WNT5a remains unknown. In this study, we generated a conditional loss-of-function mouse model (Wnt5aCAG) and examined the specific role of Wnt5a during the saccular and alveolar phases of lung development. The lack of Wnt5a in the saccular phase blocked distal airway expansion and attenuated differentiation of endothelial and alveolar epithelial type I (AT1) cells and myofibroblasts. Postnatal Wnt5a inactivation disrupted alveologenesis, producing a phenotype resembling human bronchopulmonary dysplasia (BPD). Mutant lungs showed hypoalveolization, but endothelial and epithelial differentiation was unaffected. The major impact of Wnt5a inactivation on alveologenesis was on myofibroblast differentiation and migration, with reduced expression of key regulatory genes. These findings were validated in vitro using isolated lung fibroblasts. Conditional inactivation of the WNT5a receptors Ror1 and Ror2 in alveolar myofibroblasts recapitulated the Wnt5aCAG phenotype, demonstrating that myofibroblast defects are the major cause of arrested alveologenesis in Wnt5aCAG lungs. Finally, we show that WNT5a is reduced in human BPD lung samples, indicating the clinical relevance and potential role for WNT5a in pathogenesis of BPD.
Collapse
|
28
|
Wang D, Zhang Y, Shen C. Research update on the association between SFRP5, an anti-inflammatory adipokine, with obesity, type 2 diabetes mellitus and coronary heart disease. J Cell Mol Med 2020; 24:2730-2735. [PMID: 32004418 PMCID: PMC7077606 DOI: 10.1111/jcmm.15023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/18/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Secreted frizzled-related protein 5 (SFRP5), an anti-inflammatory adipokine secreted by adipocytes, has been demonstrated to exert its anti-inflammatory effect via antagonizing the non-canonical wingless-type family member 5A (WNT5A) signalling pathways. The WNT5A protein, as a potent pro-inflammatory signalling molecule, is strongly involved in a variety of inflammatory disorders such as obesity, type 2 diabetes mellitus (T2DM) and atherosclerosis. In this review, we systematically outlined the current understanding on the roles of SFRP5 in the pathogenesis of three inflammatory diseases including obesity, T2DM and coronary heart disease (CHD). Our review might stimulate future research using SFRP5 as a promising novel therapeutic target for the treatment of obesity, T2DM and CHD.
Collapse
Affiliation(s)
- Di Wang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yaping Zhang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
29
|
Elfar N, Mohamed L, Hasby E, Kassem H. A possible role of WNT5A expression in papulosquamous skin diseases. INDIAN JOURNAL OF DERMATOPATHOLOGY AND DIAGNOSTIC DERMATOLOGY 2020. [DOI: 10.4103/ijdpdd.ijdpdd_59_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
30
|
Song F, Zou J, Song Z, Xu H, Qian Y, Zhu H, Liu S, Guan J, Chen J, Yi H. Association of Adipocytokines With Carotid Intima Media Thickness and Arterial Stiffness in Obstructive Sleep Apnea Patients. Front Endocrinol (Lausanne) 2020; 11:177. [PMID: 32300333 PMCID: PMC7142226 DOI: 10.3389/fendo.2020.00177] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/12/2020] [Indexed: 12/19/2022] Open
Abstract
Objective: Obstructive sleep apnea (OSA) results in increased carotid intima-media thickness (IMT) and arterial stiffness; however, the association between adipocytokines and IMT/arterial stiffness in OSA patients is unclear. Methods: We enrolled 95 normal weight and overweight, not obese, participants from May 2018 to December 2018 in this study. All subjects underwent a carotid artery ultrasound examination and polysomnography. Blood samples were used to determine serum chemerin, adiponectin, SFRP5, and apelin levels. Correlations between two quantitative variables were assessed using the Pearson or Spearman coefficient. Stepwise models of multiple linear regression analysis were performed to assess the independent relationships. Result: IMT in OSA patients was significantly higher than in the non-snorers. There were significant differences in the arterial stiffness parameters such as distensibility coefficient (DC), compliance coefficient (CC), and pulse wave velocity (PWV). SFRP5 level was lower in OSA patients than in non-snorers. Adiponectin correlated with CC, DC, and PWV among OSA patients; however, the relationship disappeared after a multivariable adjustment. Age was independently associated with all quantitative IMT and stiffness indices. AHI and minimum oxygen saturation (Mini SaO2) were independently related to arterial stiffness. Conclusion: The quantitative IMT and carotid arterial elasticity were significantly worse among OSA patients. Age was the main independent factor correlated with quantitative IMT and arterial stiffness, and AHI and mini SaO2 were associated factors. There were no relationships between aforementioned adipocytokines and quantitative IMT/carotid arterial stiffness.
Collapse
Affiliation(s)
- Fan Song
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Juanjuan Zou
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Zhiyuan Song
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Huajun Xu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Yinjun Qian
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Huaming Zhu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Suru Liu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Jian Guan
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Jie Chen
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- *Correspondence: Jie Chen
| | - Hongliang Yi
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Hongliang Yi
| |
Collapse
|
31
|
Endothelial Ca 2+ Signaling, Angiogenesis and Vasculogenesis: just What It Takes to Make a Blood Vessel. Int J Mol Sci 2019; 20:ijms20163962. [PMID: 31416282 PMCID: PMC6721072 DOI: 10.3390/ijms20163962] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
It has long been known that endothelial Ca2+ signals drive angiogenesis by recruiting multiple Ca2+-sensitive decoders in response to pro-angiogenic cues, such as vascular endothelial growth factor, basic fibroblast growth factor, stromal derived factor-1α and angiopoietins. Recently, it was shown that intracellular Ca2+ signaling also drives vasculogenesis by stimulation proliferation, tube formation and neovessel formation in endothelial progenitor cells. Herein, we survey how growth factors, chemokines and angiogenic modulators use endothelial Ca2+ signaling to regulate angiogenesis and vasculogenesis. The endothelial Ca2+ response to pro-angiogenic cues may adopt different waveforms, ranging from Ca2+ transients or biphasic Ca2+ signals to repetitive Ca2+ oscillations, and is mainly driven by endogenous Ca2+ release through inositol-1,4,5-trisphosphate receptors and by store-operated Ca2+ entry through Orai1 channels. Lysosomal Ca2+ release through nicotinic acid adenine dinucleotide phosphate-gated two-pore channels is, however, emerging as a crucial pro-angiogenic pathway, which sustains intracellular Ca2+ mobilization. Understanding how endothelial Ca2+ signaling regulates angiogenesis and vasculogenesis could shed light on alternative strategies to induce therapeutic angiogenesis or interfere with the aberrant vascularization featuring cancer and intraocular disorders.
Collapse
|
32
|
Circulating microRNA-378 levels serve as a novel biomarker for assessing the severity of coronary stenosis in patients with coronary artery disease. Biosci Rep 2019; 39:BSR20182016. [PMID: 31064817 PMCID: PMC6522732 DOI: 10.1042/bsr20182016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/05/2019] [Accepted: 05/02/2019] [Indexed: 01/22/2023] Open
Abstract
Background: Circulating microRNAs (miRNA) are steady preserved in blood plasma. Multiple evidences have shown that miRNAs play a crucial role in cardiovascular disease including miRNA-378, which has been illustrated to participate in diverse physiological and pathological processes of cardiovascular disease. In the present study, we aim to explore the expression of plasma miRNA-378 and its clinical significance in patients with coronary artery disease (CAD). Methods: MiRNA-378 expression in blood plasma was performed by quantitative real-time PCR (qRT-PCR) in 215 CAD patients and 52 matched controls of healthy populations. Medical information of all patients including the results of coronary angiography (CAG) was acquired through hospital information system (HIS). Spearman’s correlation, binary linear regression, and covariance analysis were used to examine the association between miRNA-378 and relative clinical risk factors. Receiver operating characteristic curve analysis was applied to evaluate the value of miRNA-378 in predicting the disease severity of coronary lesion. Results: Plasma miR-378 expression was significantly down-regulated in CAD patients compared with healthy controls. Relative miR-378 level was shown conversely correlated with Gensini score, which present the severity of coronary artery lesions. Moreover, it is indicated that miR-378 expression can effectively distinguish patients with or without coronary artery stenosis. Conclusions: Plasma miR-378 levels appear to be a promising non-invasive biomarker, but require to be further validated by a large cohort study in future.
Collapse
|
33
|
Peng S, Song C, Li H, Cao X, Ma Y, Wang X, Huang Y, Lan X, Lei C, Chaogetu B, Chen H. Circular RNA SNX29 Sponges miR-744 to Regulate Proliferation and Differentiation of Myoblasts by Activating the Wnt5a/Ca 2+ Signaling Pathway. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 16:481-493. [PMID: 31051333 PMCID: PMC6495097 DOI: 10.1016/j.omtn.2019.03.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 12/16/2022]
Abstract
Myogenesis is a complex and precisely orchestrated process that is highly regulated by several non-coding RNAs and signal pathways. Circular RNAs (circRNAs) represent a novel subclass of endogenous non-coding RNAs that have been identified in multiple species and tissues and play a vital role in post-transcriptional regulation in eukaryotes, but the precise molecular mechanism of action remains largely unknown. Here, we screened a candidate circRNA derived from the SNX29 gene, termed circSNX29 from our previous circRNAs sequencing data of bovine skeletal muscle, and further characterized its regulation and function during muscle development. The overexpression of circSNX29 facilitated myoblasts differentiation and inhibited cell proliferation. Computational analysis using RNAhybrid showed the potential for circSNX29 to sponge to miR-744 with nine potential binding sites. We tested this via a luciferase screening assay and found that circSNX29 directly interacted with miR-744 and downregulation of miR-744 efficiently reversed the suppression of Wnt5a and CaMKIIδ. Importantly, through the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enrichment analysis, Fluo-4, AM, cell permeant-calcium ion fluorescent probing, and western blotting assays, we found that overexpression of Wnt5a and circSNX29 activated the non-canonical Wnt5a/Ca2+ pathway. Overall, the evidence generated by our study elucidates the regulatory mechanisms of circSNX29 to function as a sponge for miRNA-744 in bovine primary myoblasts.
Collapse
Affiliation(s)
- Shujun Peng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Chengchuang Song
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Hui Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiukai Cao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yilei Ma
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaogang Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yongzhen Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Buren Chaogetu
- Animal Disease Control Center of Haixi Mongolian and Tibetan Autonomous Prefecture, Delingha 817000, China
| | - Hong Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
34
|
Chanda D, Otoupalova E, Smith SR, Volckaert T, De Langhe SP, Thannickal VJ. Developmental pathways in the pathogenesis of lung fibrosis. Mol Aspects Med 2018; 65:56-69. [PMID: 30130563 DOI: 10.1016/j.mam.2018.08.004] [Citation(s) in RCA: 324] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/17/2018] [Indexed: 12/20/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and terminal lung disease with no known cure. IPF is a disease of aging, with median age of diagnosis over 65 years. Median survival is between 3 and 5 years after diagnosis. IPF is characterized primarily by excessive deposition of extracellular matrix (ECM) proteins by activated lung fibroblasts and myofibroblasts, resulting in reduced gas exchange and impaired pulmonary function. Growing evidence supports the concept of a pro-fibrotic environment orchestrated by underlying factors such as genetic predisposition, chronic injury and aging, oxidative stress, and impaired regenerative responses may account for disease development and persistence. Currently, two FDA approved drugs have limited efficacy in the treatment of IPF. Many of the genes and gene networks associated with lung development are induced or activated in IPF. In this review, we analyze current knowledge in the field, gained from both basic and clinical research, to provide new insights into the disease process, and potential approaches to treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Diptiman Chanda
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Eva Otoupalova
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Samuel R Smith
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Thomas Volckaert
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Stijn P De Langhe
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
35
|
Nayak G, Odaka Y, Prasad V, Solano AF, Yeo EJ, Vemaraju S, Molkentin JD, Trumpp A, Williams B, Rao S, Lang RA. Developmental vascular regression is regulated by a Wnt/β-catenin, MYC and CDKN1A pathway that controls cell proliferation and cell death. Development 2018; 145:dev154898. [PMID: 29777010 PMCID: PMC6031408 DOI: 10.1242/dev.154898] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/08/2018] [Indexed: 12/12/2022]
Abstract
Normal development requires tight regulation of cell proliferation and cell death. Here, we have investigated these control mechanisms in the hyaloid vessels, a temporary vascular network in the mammalian eye that requires a Wnt/β-catenin response for scheduled regression. We investigated whether the hyaloid Wnt response was linked to the oncogene Myc, and the cyclin-dependent kinase inhibitor CDKN1A (P21), both established regulators of cell cycle progression and cell death. Our analysis showed that the Wnt pathway co-receptors LRP5 and LRP6 have overlapping activities that mediate the Wnt/β-catenin signaling in hyaloid vascular endothelial cells (VECs). We also showed that both Myc and Cdkn1a are downstream of the Wnt response and are required for hyaloid regression but for different reasons. Conditional deletion of Myc in VECs suppressed both proliferation and cell death. By contrast, conditional deletion of Cdkn1a resulted in VEC overproliferation that countered the effects of cell death on regression. When combined with analysis of MYC and CDKN1A protein levels, this analysis suggests that a Wnt/β-catenin and MYC-CDKN1A pathway regulates scheduled hyaloid vessel regression.
Collapse
Affiliation(s)
- Gowri Nayak
- The Visual Systems Group, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Chronobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Divisions of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yoshinobu Odaka
- The Visual Systems Group, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Chronobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Divisions of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Vikram Prasad
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Alyssa F Solano
- The Visual Systems Group, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Chronobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Divisions of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Eun-Jin Yeo
- The Visual Systems Group, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Chronobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Divisions of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Shruti Vemaraju
- The Visual Systems Group, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Chronobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Divisions of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Bart Williams
- Center for Skeletal Disease Research and Laboratory of Cell Signaling and Carcinogenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Sujata Rao
- Divisions of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- The Cleveland Clinic, Ophthalmic Research, 9500 Euclid Avenue, OH 44195, USA
| | - Richard A Lang
- The Visual Systems Group, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Chronobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Divisions of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
36
|
Padwal M, Cheng G, Liu L, Boivin F, Gangji AS, Brimble KS, Bridgewater D, Margetts PJ. WNT signaling is required for peritoneal membrane angiogenesis. Am J Physiol Renal Physiol 2018; 314:F1036-F1045. [DOI: 10.1152/ajprenal.00497.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The wingless-type mouse mammary tumor virus integration site family (WNT) signaling pathway is involved in wound healing and fibrosis. We evaluated the WNT signaling pathway in peritoneal membrane injury. We assessed WNT1 protein expression in the peritoneal effluents of 54 stable peritoneal dialysis (PD) patients and WNT-related gene expression in ex vivo mesothelial cell cultures from 21 PD patients. In a transforming growth factor-β (TGF-β)-mediated animal model of peritoneal fibrosis, we evaluated regulation of the WNT pathway and the effect of WNT inhibition on peritoneal fibrosis and angiogenesis. WNT1 and WNT2 gene expression were positively correlated with peritoneal membrane solute transport in PD patients. In the mouse peritoneum, TGF-β-induced peritoneal fibrosis was associated with increased expression of WNT2 and WNT4. Peritoneal β-catenin protein was significantly upregulated after infection with adenovirus expressing TGF-β (AdTGF-β) along with elements of the WNT signaling pathway. Treatment with a β-catenin inhibitor (ICG-001) in mice with AdTGF-β-induced peritoneal fibrosis resulted in attenuation of peritoneal angiogenesis and reduced vascular endothelial growth factor. Similar results were also observed with the WNT antagonist Dickkopf-related protein (DKK)-1. In addition to this, DKK-1 blocked epithelial-mesenchymal transition and increased levels of the cell adhesion protein E-cadherin. We provide evidence that WNT signaling is active in the setting of experimental peritoneal fibrosis and WNT1 correlates with patient peritoneal membrane solute transport in PD patients. Intervention in this pathway is a possible therapy for peritoneal membrane injury.
Collapse
Affiliation(s)
- Manreet Padwal
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Genyang Cheng
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Limin Liu
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Felix Boivin
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Azim S. Gangji
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | - Peter J. Margetts
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
37
|
Cho YK, Kang YM, Lee SE, Lee YL, Seol SM, Lee WJ, Park JY, Jung CH. Effect of SFRP5 (Secreted Frizzled-Related Protein 5) on the WNT5A (Wingless-Type Family Member 5A)-Induced Endothelial Dysfunction and Its Relevance With Arterial Stiffness in Human Subjects. Arterioscler Thromb Vasc Biol 2018; 38:1358-1367. [PMID: 29674475 DOI: 10.1161/atvbaha.117.310649] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 04/09/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE SFRP5 (secreted frizzled-related protein 5) is an endogenous inhibitor of WNT5A (wingless-type family member 5a), which has been implicated in atherosclerosis. However, contradictory results have been reported about the role of SFRP5 in atherosclerosis. We aimed to investigate whether SFRP5 could restore WNT5A-induced endothelial dysfunction in vitro and ex vivo. In addition, we sought to determine whether the serum concentration of SFRP5 is associated with atherosclerosis in humans. APPROACH AND RESULTS We measured endothelium-dependent vasorelaxation in the isolated thoracic aorta of Sprague-Dawley rats. In addition, we measured intracellular nitric oxide (NO) in human endothelial cells. The protein abundance of total and phosphorylated JNK (c-Jun N-terminal kinase), AKT (protein kinase B), and endothelial NO synthase was analyzed in human endothelial cells. Circulating SFRP5 and WNT5A levels and brachial-ankle pulse wave velocity were measured in 282 human subjects with type 2 diabetes mellitus. SFRP5 dose dependently restored Wnt5-induced impaired vasorelaxation in rat thoracic aorta by an endothelial NO synthase-dependent mechanism. SFRP5 treatment restored the WNT5A-induced reduction of NO production via endothelial NO synthase in human endothelial cells. WNT5A-induced changes in the phosphorylation of JNK, AKT, and endothelial NO synthase were ameliorated with SFRP5 administration. In humans with type 2 diabetes mellitus, the serum SFRP5 concentration positively correlated with brachial-ankle pulse wave velocity (r=0.146; P=0.024). Multivariate linear regression analysis demonstrated that the serum SFRP5 concentration was independently associated with brachial-ankle pulse wave velocity after adjustment for potential confounders [B (SE)=7.40 (3.35); P=0.028]. CONCLUSIONS Our data suggest the possible compensatory action of SFRP5 against atherosclerosis under conditions of metabolic dysfunction.
Collapse
Affiliation(s)
- Yun Kyung Cho
- From the Department of Internal Medicine (Y.K.C., Y.M.K., S.E.L., W.J.L., J.-Y.P., C.H.J.)
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea (Y.K.C.)
| | - Yu Mi Kang
- From the Department of Internal Medicine (Y.K.C., Y.M.K., S.E.L., W.J.L., J.-Y.P., C.H.J.)
| | - Seung Eun Lee
- From the Department of Internal Medicine (Y.K.C., Y.M.K., S.E.L., W.J.L., J.-Y.P., C.H.J.)
| | - Yoo La Lee
- Asan Institute of Life Sciences (Y.L.L., S.M.S.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - So Mi Seol
- Asan Institute of Life Sciences (Y.L.L., S.M.S.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo Je Lee
- From the Department of Internal Medicine (Y.K.C., Y.M.K., S.E.L., W.J.L., J.-Y.P., C.H.J.)
| | - Joong-Yeol Park
- From the Department of Internal Medicine (Y.K.C., Y.M.K., S.E.L., W.J.L., J.-Y.P., C.H.J.)
| | - Chang Hee Jung
- From the Department of Internal Medicine (Y.K.C., Y.M.K., S.E.L., W.J.L., J.-Y.P., C.H.J.)
| |
Collapse
|
38
|
The Coordinated Activities of nAChR and Wnt Signaling Regulate Intestinal Stem Cell Function in Mice. Int J Mol Sci 2018; 19:ijms19030738. [PMID: 29510587 PMCID: PMC5877599 DOI: 10.3390/ijms19030738] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 02/27/2018] [Accepted: 03/03/2018] [Indexed: 12/14/2022] Open
Abstract
Cholinergic signaling, which modulates cell activities via nicotinic and muscarinic acetylcholine receptors (n- and mAChRs) in response to internal or external stimuli, has been demonstrated in mammalian non-neuronal cells that synthesize acetylcholine (ACh). One of the major pathways of excitatory transmission in the enteric nervous system (ENS) is mediated by cholinergic transmission, with the transmitter ACh producing excitatory potentials in postsynaptic effector cells. In addition to ACh-synthesizing and ACh-metabolizing elements in the ENS, the presence of non-neuronal ACh machinery has been reported in epithelial cells of the small and large intestines of rats and humans. However, little is known about how non-neuronal ACh controls physiological function in the intestine. Here, experiments using crypt-villus organoids that lack nerve and immune cells in culture suggest that endogenous ACh is synthesized in the intestinal epithelium to drive organoid growth and differentiation through activation of nAChRs. Treatment of organoids with nicotine enhanced cell growth and the expression of marker genes for stem and epithelial cells. On the other hand, the nAChR antagonist mecamylamine strongly inhibited the growth and differentiation of organoids, suggesting the involvement of nAChRs in the regulation of proliferation and differentiation of Lgr5-positive stem cells. More specifically, RNA sequencing analysis revealed that Wnt5a expression was dramatically upregulated after nicotine treatment, and Wnt5a rescued organoid growth and differentiation in response to mecamylamine. Taken together, our results indicate that coordinated activities of nAChR and Wnt signaling maintain Lgr5-positive stem cell activity and balanced differentiation. Furthermore, we could clearly separate the two groups, neuronal ACh in the ENS and non-neuronal ACh in the intestinal epithelium. Dysfunction of the non-neuronal cholinergic system is involved in the pathogenesis of disease. The data will increase our understanding of the cholinergic properties of non-neuronal cells and lead to optimization of drug therapy.
Collapse
|
39
|
Zeng C, Guo C, Cai J, Tang C, Dong Z. Serum sclerostin in vascular calcification and clinical outcome in chronic kidney disease. Diab Vasc Dis Res 2018; 15:99-105. [PMID: 29168393 DOI: 10.1177/1479164117742316] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Sclerostin, a potent soluble inhibitor of the Wnt signalling pathway, is known to inhibit bone formation by suppressing osteocytes differentiation and function. Patients with chronic kidney disease have high levels of serum sclerostin. Sclerostin has been implicated in the pathogenesis of vascular calcification, which may promote the cardiovascular events of morbidity and mortality in chronic kidney disease patients. However, the role of sclerostin in vascular calcification and clinical prognosis in chronic kidney disease remains elusive. While some studies suggested a positive correlation between serum sclerostin and vascular calcification or clinical outcome, other studies showed no or even negative correlation between them. Small sample size, heterogeneity in enrolled patients, discrepancy in anatomical structure examined and differences in the applied assays may be responsible for the discrepant results. Nonetheless, anti-sclerostin antibodies may be a new therapeutic approach to increase bone mass and strength in chronic kidney disease. This review aims to have a better understanding of the relationship of serum sclerostin with vascular calcification and clinical outcome in chronic kidney disease patients, and propose the application of anti-sclerostin therapy in chronic kidney disease.
Collapse
Affiliation(s)
- Cong Zeng
- 1 Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chunyuan Guo
- 2 Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Juan Cai
- 1 Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chengyuan Tang
- 1 Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zheng Dong
- 1 Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- 2 Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| |
Collapse
|
40
|
Shao Y, Zheng Q, Wang W, Xin N, Song X, Zhao C. Biological functions of macrophage-derived Wnt5a, and its roles in human diseases. Oncotarget 2018; 7:67674-67684. [PMID: 27608847 PMCID: PMC5341904 DOI: 10.18632/oncotarget.11874] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 08/27/2016] [Indexed: 12/31/2022] Open
Abstract
Wnt5a is implicated in development and tissue homeostasis by activating β-catenin-independent pathway. Excessive production of Wnt5a is related to some human diseases. Macrophage recruitment is a character of inflammation and cancer, therefore macrophage-derived Wnt5a is supposed to be a player in these conditions. Actually, macrophage-derived Wnt5a maintains macrophage immune function, stimulates pro-inflammatory cytokine release, and induces angiogenesis and lymphangiogenesis. Furthermore, macrophage-derived Wnt5a is involved in insulin resistance, atherosclerosis and cancer. These findings indicate that macrophage-derived Wnt5a may be a target in the treatment of these diseases. Notably, unlike macrophages, the exact role of macrophage-derived Wnt5a in bacterial infection remains largely unknown.
Collapse
Affiliation(s)
- Yue Shao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Qianqian Zheng
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Na Xin
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Xiaowen Song
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
41
|
Mehta S, Lo Cascio C. Developmentally regulated signaling pathways in glioma invasion. Cell Mol Life Sci 2018; 75:385-402. [PMID: 28821904 PMCID: PMC5765207 DOI: 10.1007/s00018-017-2608-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/18/2017] [Accepted: 08/03/2017] [Indexed: 01/06/2023]
Abstract
Malignant gliomas are the most common, infiltrative, and lethal primary brain tumors affecting the adult population. The grim prognosis for this disease is due to a combination of the presence of highly invasive tumor cells that escape surgical resection and the presence of a population of therapy-resistant cancer stem cells found within these tumors. Several studies suggest that glioma cells have cleverly hijacked the normal developmental program of neural progenitor cells, including their transcriptional programs, to enhance gliomagenesis. In this review, we summarize the role of developmentally regulated signaling pathways that have been found to facilitate glioma growth and invasion. Furthermore, we discuss how the microenvironment and treatment-induced perturbations of these highly interconnected signaling networks can trigger a shift in cellular phenotype and tumor subtype.
Collapse
Affiliation(s)
- Shwetal Mehta
- Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, 85013, USA.
| | - Costanza Lo Cascio
- Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, 85013, USA
| |
Collapse
|
42
|
Foulquier S, Daskalopoulos EP, Lluri G, Hermans KCM, Deb A, Blankesteijn WM. WNT Signaling in Cardiac and Vascular Disease. Pharmacol Rev 2018; 70:68-141. [PMID: 29247129 PMCID: PMC6040091 DOI: 10.1124/pr.117.013896] [Citation(s) in RCA: 258] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
WNT signaling is an elaborate and complex collection of signal transduction pathways mediated by multiple signaling molecules. WNT signaling is critically important for developmental processes, including cell proliferation, differentiation and tissue patterning. Little WNT signaling activity is present in the cardiovascular system of healthy adults, but reactivation of the pathway is observed in many pathologies of heart and blood vessels. The high prevalence of these pathologies and their significant contribution to human disease burden has raised interest in WNT signaling as a potential target for therapeutic intervention. In this review, we first will focus on the constituents of the pathway and their regulation and the different signaling routes. Subsequently, the role of WNT signaling in cardiovascular development is addressed, followed by a detailed discussion of its involvement in vascular and cardiac disease. After highlighting the crosstalk between WNT, transforming growth factor-β and angiotensin II signaling, and the emerging role of WNT signaling in the regulation of stem cells, we provide an overview of drugs targeting the pathway at different levels. From the combined studies we conclude that, despite the sometimes conflicting experimental data, a general picture is emerging that excessive stimulation of WNT signaling adversely affects cardiovascular pathology. The rapidly increasing collection of drugs interfering at different levels of WNT signaling will allow the evaluation of therapeutic interventions in the pathway in relevant animal models of cardiovascular diseases and eventually in patients in the near future, translating the outcomes of the many preclinical studies into a clinically relevant context.
Collapse
Affiliation(s)
- Sébastien Foulquier
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Evangelos P Daskalopoulos
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Gentian Lluri
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Arjun Deb
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| |
Collapse
|
43
|
Shi YN, Zhu N, Liu C, Wu HT, Gui Y, Liao DF, Qin L. Wnt5a and its signaling pathway in angiogenesis. Clin Chim Acta 2017. [DOI: 10.1016/j.cca.2017.06.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
44
|
Hu B, Wang Q, Wang YA, Hua S, Sauvé CEG, Ong D, Lan ZD, Chang Q, Ho YW, Monasterio MM, Lu X, Zhong Y, Zhang J, Deng P, Tan Z, Wang G, Liao WT, Corley LJ, Yan H, Zhang J, You Y, Liu N, Cai L, Finocchiaro G, Phillips JJ, Berger MS, Spring DJ, Hu J, Sulman EP, Fuller GN, Chin L, Verhaak RGW, DePinho RA. Epigenetic Activation of WNT5A Drives Glioblastoma Stem Cell Differentiation and Invasive Growth. Cell 2017; 167:1281-1295.e18. [PMID: 27863244 DOI: 10.1016/j.cell.2016.10.039] [Citation(s) in RCA: 204] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/11/2016] [Accepted: 10/20/2016] [Indexed: 01/08/2023]
Abstract
Glioblastoma stem cells (GSCs) are implicated in tumor neovascularization, invasiveness, and therapeutic resistance. To illuminate mechanisms governing these hallmark features, we developed a de novo glioblastoma multiforme (GBM) model derived from immortalized human neural stem/progenitor cells (hNSCs) to enable precise system-level comparisons of pre-malignant and oncogene-induced malignant states of NSCs. Integrated transcriptomic and epigenomic analyses uncovered a PAX6/DLX5 transcriptional program driving WNT5A-mediated GSC differentiation into endothelial-like cells (GdECs). GdECs recruit existing endothelial cells to promote peritumoral satellite lesions, which serve as a niche supporting the growth of invasive glioma cells away from the primary tumor. Clinical data reveal higher WNT5A and GdECs expression in peritumoral and recurrent GBMs relative to matched intratumoral and primary GBMs, respectively, supporting WNT5A-mediated GSC differentiation and invasive growth in disease recurrence. Thus, the PAX6/DLX5-WNT5A axis governs the diffuse spread of glioma cells throughout the brain parenchyma, contributing to the lethality of GBM.
Collapse
Affiliation(s)
- Baoli Hu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qianghu Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Y Alan Wang
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Sujun Hua
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Derrick Ong
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zheng D Lan
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qing Chang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Wing Ho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marta Moreno Monasterio
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xin Lu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yi Zhong
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pingna Deng
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhi Tan
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guocan Wang
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wen-Ting Liao
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lynda J Corley
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Haiyan Yan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ning Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Linbo Cai
- Department of Oncology, Guangdong 999 Brain Hospital, Guangzhou 510510, China
| | - Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico C. Besta, 20133 Milano, Italy
| | - Joanna J Phillips
- Departments of Neurological Surgery and Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mitchel S Berger
- Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Denise J Spring
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Hu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Erik P Sulman
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gregory N Fuller
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lynda Chin
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Roeland G W Verhaak
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
45
|
Ahsan R, Baisiwala S, Ahmed AU. Rogue one: another faction of the Wnt empire implicated in assisting GBM progression. Transl Cancer Res 2017; 6:S321-S327. [PMID: 30662831 DOI: 10.21037/tcr.2017.03.29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
It remains incumbent on researchers to conceive novel treatments for the most common primary malignancy of the brain in adults, glioblastoma multiforme (GBM), as the standard of care for patients today fails to yield a median survival beyond two years following diagnosis. Recent studies have tended towards appreciating the cellular heterogeneity of GBM tumors, focusing on the subpopulation of highly plastic glioblastoma stem cells (GSCs). In the November 2016 issue of Cell, Hu and colleagues developed a de nova GBM model derived from immortalized neural stem cells and, using this model, they demonstrated that GSCs can generate CD133+/CD144+ cells with endothelial cell-like characteristics. Contrasts between the epigenetic state and gene expression level before and after oncogenic transformation of this utilized de novo model for GBM implicated WNT5A, which has been previously shown to play a role in endothelial cell proliferation and migration via non-canonical Wnt signaling, as a mediator of the process. The transdifferentiation was accompanied by alterations in the histone marks at the gene loci of WNT5A, and its transcription factors PAX6 and DXL5. The authors hypothesize that activation of AKT, an aberration of the RTK/PTEN/PI3K pathway observed in the majority of GBM cases, triggers these epigenetic changes causing WNT5A expression. This phenomenon is of obvious clinical significance, as it provides an insight into how GBM may circumvent therapies targeting angiogenesis to achieve the neovascularization required to sustain invasive growth. The unveiling of this atypical differentiation process also raises questions about its interaction with the radiotherapy and chemotherapy commonly used to counter GBM progression. Here, we review the recent efforts to understand the complex mechanisms behind the plasticity of GSCs.
Collapse
Affiliation(s)
- Riasat Ahsan
- The Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Shivani Baisiwala
- The Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Atique U Ahmed
- The Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
46
|
Pashirzad M, Shafiee M, Rahmani F, Behnam-Rassouli R, Hoseinkhani F, Ryzhikov M, Moradi Binabaj M, Parizadeh MR, Avan A, Hassanian SM. Role of Wnt5a in the Pathogenesis of Inflammatory Diseases. J Cell Physiol 2017; 232:1611-1616. [DOI: 10.1002/jcp.25687] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/09/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Mehran Pashirzad
- Department of Medical Biochemistry; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mojtaba Shafiee
- Department of Nutrition; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Farzad Rahmani
- Department of Medical Biochemistry; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | | | - Fatemeh Hoseinkhani
- Department of Medical Biochemistry; School of Medicine; Iran University of Medical Sciences; Tehran Iran
| | - Mikhail Ryzhikov
- Department of Biochemistry and Molecular Biology; St. Louis University School of Medicine; Saint Louis Missouri
| | - Maryam Moradi Binabaj
- Department of Medical Biochemistry; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mohammad Reza Parizadeh
- Department of Medical Biochemistry; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Amir Avan
- Molecular Medicine Group; Department of Modern Sciences and Technologies; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Departments of Medical Oncology and Neurology; VU University Medical Center; Amsterdam The Netherlands
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Department of Biochemistry and Molecular Biology; St. Louis University School of Medicine; Saint Louis Missouri
- Microanatomy Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- Metabolic Syndrome Research Center; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
47
|
Rapp J, Kiss E, Meggyes M, Szabo-Meleg E, Feller D, Smuk G, Laszlo T, Sarosi V, Molnar TF, Kvell K, Pongracz JE. Increased Wnt5a in squamous cell lung carcinoma inhibits endothelial cell motility. BMC Cancer 2016; 16:915. [PMID: 27876017 PMCID: PMC5120464 DOI: 10.1186/s12885-016-2943-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/09/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Angiogenesis is important both in normal tissue function and disease and represents a key target in lung cancer (LC) therapy. Unfortunately, the two main subtypes of non-small-cell lung cancers (NSCLC) namely, adenocarcinoma (AC) and squamous cell carcinoma (SCC) respond differently to anti-angiogenic e.g. anti-vascular endothelial growth factor (VEGF)-A treatment with life-threatening side effects, often pulmonary hemorrhage in SCC. The mechanisms behind such adverse reactions are still largely unknown, although peroxisome proliferator activator receptor (PPAR) gamma as well as Wnt-s have been named as molecular regulators of the process. As the Wnt microenvironments in NSCLC subtypes are drastically different, we hypothesized that the particularly high levels of non-canonical Wnt5a in SCC might be responsible for alterations in blood vessel growth and result in serious adverse reactions. METHODS PPARgamma, VEGF-A, Wnt5a, miR-27b and miR-200b levels were determined in resected adenocarcinoma and squamous cell carcinoma samples by qRT-PCR and TaqMan microRNA assay. The role of PPARgamma in VEGF-A expression, and the role of Wnts in overall regulation was investigated using PPARgamma knock-out mice, cancer cell lines and fully human, in vitro 3 dimensional (3D), distal lung tissue aggregates. PPARgamma mRNA and protein levels were tested by qRT-PCR and immunohistochemistry, respectively. PPARgamma activity was measured by a PPRE reporter system. The tissue engineered lung tissues expressing basal level and lentivirally delivered VEGF-A were treated with recombinant Wnts, chemical Wnt pathway modifiers, and were subjected to PPARgamma agonist and antagonist treatment. RESULTS PPARgamma down-regulation and VEGF-A up-regulation are characteristic to both AC and SCC. Increased VEGF-A levels are under direct control of PPARgamma. PPARgamma levels and activity, however, are under Wnt control. Imbalance of both canonical (in AC) and non-canonical (in SCC) Wnts leads to PPARgamma down-regulation. While canonical Wnts down-regulate PPARgamma directly, non-canonical Wnt5a increases miR27b that is known regulator of PPARgamma. CONCLUSION During carcinogenesis the Wnt microenvironment alters, which can downregulate PPARgamma leading to increased VEGF-A expression. Differences in the Wnt microenvironment in AC and SCC of NSCLC lead to PPARgamma decrease via mechanisms that differentially alter endothelial cell motility and branching which in turn can influence therapeutic response.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Animals
- Biomarkers, Tumor
- Carcinoma, Non-Small-Cell Lung/blood supply
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Movement
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/blood supply
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- PPAR gamma/physiology
- Tumor Cells, Cultured
- Tumor Microenvironment
- Vascular Endothelial Growth Factor A/metabolism
- Wnt-5a Protein/metabolism
Collapse
Affiliation(s)
- J Rapp
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary
| | - E Kiss
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary
| | - M Meggyes
- Medical Microbiology and Immunity, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary
| | - E Szabo-Meleg
- Biophysics, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
| | - D Feller
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary
| | - G Smuk
- Pathology, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
| | - T Laszlo
- Pathology, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
| | - V Sarosi
- Internal Medicine, Pulmonology, University of Pécs, 2 Rakoczi Str, Pécs, 7623, Hungary
| | - T F Molnar
- Operational Medicine, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
- Department of Surgery, Thoracic Surgery Unit, Petz A Hospital, 2-4 Vasvari Str, Győr, 9023, Hungary
| | - K Kvell
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
| | - J E Pongracz
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary.
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary.
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary.
| |
Collapse
|
48
|
Wang SH, Chang JS, Hsiao JR, Yen YC, Jiang SS, Liu SH, Chen YL, Shen YY, Chang JY, Chen YW. Tumour cell-derived WNT5B modulates in vitro lymphangiogenesis via induction of partial endothelial-mesenchymal transition of lymphatic endothelial cells. Oncogene 2016; 36:1503-1515. [PMID: 27593938 DOI: 10.1038/onc.2016.317] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/03/2016] [Accepted: 07/25/2016] [Indexed: 12/22/2022]
Abstract
Metastasis of the cervical lymph nodes frequently leads to poor survival of patients with oral squamous cell carcinoma (OSCC). The underlying mechanisms of lymph node metastasis are unclear. Wingless-type MMTV integration site family, member 5B (WNT5B), one component of the WNT signal pathway, was markedly up-regulated in OSCC sublines with high potential of lymphatic metastasis compared to that in OSCC cells with low nodal metastasis. Increased WNT5B mRNA was demonstrated in human OSCC tissues in comparison with adjacent non-tumorous tissues. Interestingly, the high level of WNT5B protein in serum was associated with lymph node metastasis in OSCC patients. Knockdown of WNT5B expression in OSCC sublines did not affect tumour growth but impaired lymph node metastasis and tumour lymphangiogenesis of orthotopic transplantation. Conditioned medium from WNT5B knockdown cells reduced the tube formation of lymphatic endothelial cells (LECs). In contrast, recombinant WNT5B enhanced the tube formation, permeability and migration of LECs. In LECs stained with phalloidin, the morphology of those treated with recombinant WNT5B changed from flat to spindle-like. Recombinant WNT5B also increased α-smooth muscle actin and inhibited the expression of vascular endothelial-cadherin but retained characteristics of endothelial cells. The results suggest that WNT5B functions in the partial endothelial-mesenchymal transition (EndoMT). Furthermore, WNT5B-induced tube formation was impaired in the LECs following the knockdown of EndoMT-related transcription factor, SNAIL or SLUG. The WNT5B-induced expression of Snail or Slug was abolished by IWR-1-endo and Rac1 inhibitors, which are involved in the WNT/β-catenin and planar cell polarity pathways, respectively. Collectively, the data suggest that WNT5B induces tube formation by regulating the expression of Snail and Slug proteins through activation of canonical and non-canonical WNT signalling pathways.
Collapse
Affiliation(s)
- S-H Wang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - J S Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - J-R Hsiao
- Department of Otolaryngology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Y-C Yen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - S S Jiang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - S-H Liu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Y-L Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Y-Y Shen
- Pathology Core Laboratory, National Health Research Institutes, Miaoli, Taiwan
| | - J-Y Chang
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Y-W Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan.,PhD Program for Aging, Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
49
|
Gerhardt C, Leu T, Lier JM, Rüther U. The cilia-regulated proteasome and its role in the development of ciliopathies and cancer. Cilia 2016; 5:14. [PMID: 27293550 PMCID: PMC4901515 DOI: 10.1186/s13630-016-0035-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/29/2016] [Indexed: 12/21/2022] Open
Abstract
The primary cilium is an essential structure for the mediation of numerous signaling pathways involved in the coordination and regulation of cellular processes essential for the development and maintenance of health. Consequently, ciliary dysfunction results in severe human diseases called ciliopathies. Since many of the cilia-mediated signaling pathways are oncogenic pathways, cilia are linked to cancer. Recent studies demonstrate the existence of a cilia-regulated proteasome and that this proteasome is involved in cancer development via the progression of oncogenic, cilia-mediated signaling. This review article investigates the association between primary cilia and cancer with particular emphasis on the role of the cilia-regulated proteasome.
Collapse
Affiliation(s)
- Christoph Gerhardt
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Tristan Leu
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Johanna Maria Lier
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Ulrich Rüther
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
50
|
Haxaire C, Haÿ E, Geoffroy V. Runx2 Controls Bone Resorption through the Down-Regulation of the Wnt Pathway in Osteoblasts. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1598-609. [DOI: 10.1016/j.ajpath.2016.01.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 12/21/2015] [Accepted: 01/21/2016] [Indexed: 12/27/2022]
|