201
|
Turk JR, Deaton AM, Yin J, Stolina M, Felx M, Boyd G, Bienvenu JG, Varela A, Guillot M, Holdsworth G, Wolfreys A, Dwyer D, Kumar SV, de Koning EM, Qu Y, Engwall M, Locher K, Ward LD, Glaus C, He YD, Boyce RW. Nonclinical cardiovascular safety evaluation of romosozumab, an inhibitor of sclerostin for the treatment of osteoporosis in postmenopausal women at high risk of fracture. Regul Toxicol Pharmacol 2020; 115:104697. [PMID: 32590049 DOI: 10.1016/j.yrtph.2020.104697] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/20/2020] [Accepted: 05/29/2020] [Indexed: 12/26/2022]
Abstract
Romosozumab (EVENITY™ [romosozumab-aqqg in the US]) is a humanized monoclonal antibody that inhibits sclerostin and has been approved in several countries for the treatment of osteoporosis in postmenopausal women at high risk of fracture. Sclerostin is expressed in bone and aortic vascular smooth muscle (AVSM). Its function in AVSM is unclear but it has been proposed to inhibit vascular calcification, atheroprogression, and inflammation. An increased incidence of positively adjudicated serious cardiovascular adverse events driven by an increase in myocardial infarction and stroke was observed in romosozumab-treated subjects in a clinical trial comparing alendronate with romosozumab (ARCH; NCT01631214) but not in a placebo-controlled trial (FRAME; NCT01575834). To investigate the effects of sclerostin inhibition with sclerostin antibody on the cardiovascular system, a comprehensive nonclinical toxicology package with additional cardiovascular studies was conducted. Although pharmacodynamic effects were observed in the bone, there were no functional, morphological, or transcriptional effects on the cardiovascular system in animal models in the presence or absence of atherosclerosis. These nonclinical studies did not identify evidence that proves the association between sclerostin inhibition and adverse cardiovascular function, increased cardiovascular calcification, and atheroprogression.
Collapse
Affiliation(s)
- James R Turk
- Translational Safety and Bioanalytical Sciences, Amgen Research, Thousand Oaks, CA, USA.
| | - Aimee M Deaton
- Translational Safety and Bioanalytical Sciences, Amgen Research, Cambridge, MA, USA
| | - Jun Yin
- Genome Analysis Unit, Amgen Research, South San Francisco, CA, USA
| | - Marina Stolina
- Cardiometabolic Disorders Research, Amgen Research, Thousand Oaks, CA, USA
| | - Melanie Felx
- Charles River Laboratories Montreal ULC, Senneville, QC, Canada
| | - Gabrielle Boyd
- Charles River Laboratories Montreal ULC, Senneville, QC, Canada
| | | | - Aurore Varela
- Charles River Laboratories Montreal ULC, Senneville, QC, Canada
| | - Martin Guillot
- Charles River Laboratories Montreal ULC, Senneville, QC, Canada
| | | | | | - Denise Dwyer
- Cardiometabolic Disorders Research, Amgen Research, Thousand Oaks, CA, USA
| | - Sheetal V Kumar
- Translational Safety and Bioanalytical Sciences, Amgen Research, Cambridge, MA, USA
| | - Emily M de Koning
- Translational Safety and Bioanalytical Sciences, Amgen Research, Cambridge, MA, USA
| | - Yusheng Qu
- Translational Safety and Bioanalytical Sciences, Amgen Research, Thousand Oaks, CA, USA
| | - Michael Engwall
- Translational Safety and Bioanalytical Sciences, Amgen Research, Thousand Oaks, CA, USA
| | - Kathrin Locher
- Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Lucas D Ward
- Translational Safety and Bioanalytical Sciences, Amgen Research, Cambridge, MA, USA
| | - Charles Glaus
- Cardiometabolic Disorders Research, Amgen Research, Thousand Oaks, CA, USA
| | - Yudong D He
- Translational Safety and Bioanalytical Sciences, Amgen Research, Thousand Oaks, CA, USA; Genome Analysis Unit, Amgen Research, South San Francisco, CA, USA
| | - Rogely Waite Boyce
- Translational Safety and Bioanalytical Sciences, Amgen Research, Thousand Oaks, CA, USA
| |
Collapse
|
202
|
Neuro-transcriptomic signatures for mood disorder morbidity and suicide mortality. J Psychiatr Res 2020; 127:62-74. [PMID: 32485434 DOI: 10.1016/j.jpsychires.2020.05.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 03/30/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
Suicidal behaviors are strongly linked with mood disorders, but the specific neurobiological and functional gene-expression correlates for this linkage remain elusive. We performed neuroimaging-guided RNA-sequencing in two studies to test the hypothesis that imaging-localized gray matter volume (GMV) loss in mood disorders, harbors gene-expression changes associated with disease morbidity and related suicide mortality in an independent postmortem cohort. To do so, first, we conducted study 1 using an anatomical likelihood estimation (ALE) MRI meta-analysis including a total of 47 voxel-based morphometry (VBM) publications (i.e. 26 control versus (vs) major depressive disorder (MDD) studies, and 21 control vs bipolar disorder (BD) studies) in 2387 (living) participants. Study 1 meta-analysis identified a selective anterior insula cortex (AIC) GMV loss in mood disorders. We then used this results to guide study 2 postmortem tissue dissection and RNA-Sequencing of 100 independent donor brain samples with a life-time history of MDD (N = 30), BD (N = 37) and control (N = 33). In study 2, exploratory factor-analysis identified a higher-order factor representing number of Axis-1 diagnoses (e.g. substance use disorders/psychosis/anxiety, etc.), referred to here as morbidity and suicide-completion referred to as mortality. Comparisons of case-vs-control, and factor-analysis defined higher-order-factor contrast variables revealed that the imaging-identified AIC GMV loss sub-region harbors differential gene-expression changes in high morbidity-&-mortality versus low morbidity-&-mortality cohorts in immune, inflammasome, and neurodevelopmental pathways. Weighted gene co-expression network analysis further identified co-activated gene modules for psychiatric morbidity and mortality outcomes. These results provide evidence that AIC anatomical signature for mood disorders are possible correlates for gene-expression abnormalities in mood morbidity and suicide mortality.
Collapse
|
203
|
Javed Z, Khan K, Sadia H, Raza S, Salehi B, Sharifi-Rad J, Cho WC. LncRNA & Wnt signaling in colorectal cancer. Cancer Cell Int 2020; 20:326. [PMID: 32699525 PMCID: PMC7372757 DOI: 10.1186/s12935-020-01412-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
The outlook for new therapeutic approaches is pivotal to ameliorate the deterioration caused by the abrogated Wnt signaling. Long non-coding RNAs (lncRNAs) are tiny molecules that have begun emerging as vital molecular manager for the regulation of various cellular processes at transcription and translation levels in the colorectal cancer (CRC). Targeting Wnt pathway with lncRNA seems a promising approach to eradicate CRC. However, little is known of their active role in commencing both apoptosis and proliferation in CRC. This article reviews the importance of these molecules in the pathogenesis of CRC and also emphasizes on the development of new therapeutic strategies to cope with the Wnt mediated CRC.
Collapse
Affiliation(s)
- Zeeshan Javed
- Office for Research Innovation and Commercialization, Lahore Garrison University, Sector-C, Phase VI, DHA, Lahore, Pakistan
| | - Khushbukhat Khan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000 Pakistan
| | - Haleema Sadia
- Department of Biotechnology, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, Pakistan
| | - Shahid Raza
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan
| | - Bahare Salehi
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong, China
| |
Collapse
|
204
|
Serafino A, Giovannini D, Rossi S, Cozzolino M. Targeting the Wnt/β-catenin pathway in neurodegenerative diseases: recent approaches and current challenges. Expert Opin Drug Discov 2020; 15:803-822. [PMID: 32281421 DOI: 10.1080/17460441.2020.1746266] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/19/2020] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Wnt/β-catenin signaling is an evolutionarily conserved pathway having a crucial role in embryonic and adult life. Specifically, the Wnt/β-catenin axis is pivotal to the development and homeostasis of the nervous system, and its dysregulation has been associated with various neurological disorders, including neurodegenerative diseases. Therefore, this signaling pathway has been proposed as a potential therapeutic target against neurodegeneration. AREAS COVERED This review focuses on the role of Wnt/β-catenin pathway in the pathogenesis of neurodegenerative diseases, including Parkinson's, Alzheimer's Diseases and Amyotrophic Lateral Sclerosis. The evidence showing that defects in the signaling might be involved in the development of these diseases, and the pharmacological approaches tested so far, are discussed. The possibilities that this pathway offers in terms of new therapeutic opportunities are also considered. EXPERT OPINION The increasing interest paid to the role of Wnt/β-catenin pathway in the onset of neurodegenerative diseases demonstrates how targeting this signaling for therapeutic purposes could be a great opportunity for both neuroprotection and neurorepair. Without overlooking some licit concerns about drug safety and delivery to the brain, there is growing and more convincing evidence that restoring this signaling in neurodegenerative diseases may strongly increase the chance to develop disease-modifying treatments for these brain pathologies.
Collapse
Affiliation(s)
- Annalucia Serafino
- Institute of Translational Pharmacology, National Research Council (CNR) , Rome, Italy
| | - Daniela Giovannini
- Institute of Translational Pharmacology, National Research Council (CNR) , Rome, Italy
| | - Simona Rossi
- Institute of Translational Pharmacology, National Research Council (CNR) , Rome, Italy
| | - Mauro Cozzolino
- Institute of Translational Pharmacology, National Research Council (CNR) , Rome, Italy
| |
Collapse
|
205
|
Arisan ED, Rencuzogullari O, Keskin B, Grant GH, Uysal-Onganer P. Inhibition on JNK Mimics Silencing of Wnt-11 Mediated Cellular Response in Androgen-Independent Prostate Cancer Cells. BIOLOGY 2020; 9:biology9070142. [PMID: 32605008 PMCID: PMC7407974 DOI: 10.3390/biology9070142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 11/23/2022]
Abstract
Prostate cancer (PCa) is one of the most common cancers among men, and one of the leading causes of cancer death for men. The c-Jun N-terminal kinase (JNK) pathway is required for several cellular functions, such as survival, proliferation, differentiation, and migration. Wnt-11, a member of the Wnt family, has been identified for its upregulation in PCa; however, downstream signalling of Wnt-11 remains to be fully characterized. In this study, we investigated the role of the JNK pathway as a potential downstream factor for Wnt-11 signalling. For this purpose, LNCaP, DU145, and PC-3 PCa cells and normal epithelial PNT1A cells were treated with a specific JNK kinase inhibitor: JNKVIII. Our results showed that JNK inhibition decreased mitochondrial membrane potential and promoted cell death in a cell type-dependent manner. We found that JNK inhibition led to an increase in autophagy and prevented epithelial–mesenchymal transition (EMT) in independently growing androgen cells. JNK inhibition and the silencing of Wnt-11 showed similar responses in DU145 and PC-3 cells and decreased metastasis-related biomarkers, cell migration, and invasion. Overall, our results suggest that JNK signalling plays a significant role in the pathophysiology of PCa by mediating Wnt-11 induced signals. Our data highlights that both the JNK pathway and Wnt-11 could be a useful therapeutic target for the combinatory application of current PCa.
Collapse
Affiliation(s)
- Elif Damla Arisan
- Gebze Technical University, Institute of Biotechnology, 41400 Gebze-Kocaeli, Turkey;
| | - Ozge Rencuzogullari
- Istanbul Kultur University, Department of Molecular Biology and Genetics, Atakoy Campus, 34156 Istanbul, Turkey; (O.R.); (B.K.)
| | - Buse Keskin
- Istanbul Kultur University, Department of Molecular Biology and Genetics, Atakoy Campus, 34156 Istanbul, Turkey; (O.R.); (B.K.)
| | - Guy H. Grant
- School of Life Sciences, University of Bedfordshire, Park Square, Luton LU1 3JU, UK;
| | - Pinar Uysal-Onganer
- Cancer Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK
- Correspondence: ; Tel.: +44-(0)207-911-5151 (ext. 64581)
| |
Collapse
|
206
|
Shin J, Yoon Y, Oh DJ. Evaluation of the Wnt signaling pathway as a prognostic marker in patients with urosepsis. Mol Cell Biochem 2020; 473:15-23. [PMID: 32588279 DOI: 10.1007/s11010-020-03804-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/18/2020] [Indexed: 12/30/2022]
Abstract
The Wnt signaling pathway has critical roles in dysregulated inflammation during sepsis; however, its impacts on clinical outcomes remain uncertain. This prospective observational study investigated the association between the Wnt pathway and clinical outcomes in patients with urosepsis. The study included 38 patients with urosepsis and 20 healthy individuals. Wnt3a and Wnt5a levels were measured at admission. The primary outcome was the occurrence of major adverse kidney events (MAKE), defined as new renal replacement therapy, stage 3 acute kidney injury, or death. Both Wnt3a and Wnt5a levels were higher in the patient group than in the control (P = 0.001 and P < 0.001, respectively). The primary outcome occurred in 13 (34.2%) subjects. The levels of Wnt5a were higher in subjects with MAKE than in those without MAKE (P = 0.015); however, Wnt3a levels showed no significant difference. Moreover, Wnt5a levels could be a marker to predict the possibility of MAKE (area under the curve 0.74 [0.57-0.92]; P = 0.016). Serum creatinine levels on day 0, day 5, and on discharge day were evaluated. The levels of creatinine on discharge day were higher in patients with high Wnt5a levels, compared to those with low Wnt5a levels (P = 0.030); however, no difference in Wnt5a levels was observed on day 0 and 5. Wnt3a and Wnt5a levels increased in patients with urosepsis. Moreover, evaluation of Wnt5a levels might help to predict the occurrence of MAKE and renal recovery in these patients.
Collapse
Affiliation(s)
- Jungho Shin
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Yoosik Yoon
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Dong-Jin Oh
- Department of Internal Medicine, Myongji Hospital, Hanyang University College of Medicine, 5 Hwasu-ro 14, Deogyang-gu, Goyang, 10475, Korea.
| |
Collapse
|
207
|
Tepekoy F, Akkoyunlu G. The interaction of Wnt signaling members with growth factors in cultured granulosa cells. Anim Reprod 2020; 17:e20190106. [PMID: 32714449 PMCID: PMC7375871 DOI: 10.1590/1984-3143-ar2019-0106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Wnt family members have recently been distinguished in the adult ovary with potential roles in ovarian function. Though particular growth factors interact with Wnt signaling members in extraovarian cell types, it is unclear whether this interaction is applicable in the granulosa cells. Therefore, the current study aimed to determine the effect of insulin-like growth factor-1 (IGF-I), epidermal growth factor (EGF) and basic fibroblast growth factor (FGF-β) on Wnt ligands WNT2 and WNT4 and Wnt receptor Frizzled-4 (FZD4) protein levels in cultured mouse granulosa cells. Granulosa cells were isolated from antral follicles of adult Balb/C mice and cultured for 24 hours in the presence of 100 ng/mL of IGF-I, or EGF or FGF-β. WNT2, WNT4 and FZD4 protein levels were evaluated through western blotting after the culture process. IGF-I treated granulosa cells had significantly the highest level of WNT2 and WNT4 as well as FZD4 when compared to FGF-β and EGF groups. FGF-β group had a significantly higher level of WNT2, WNT4 and FZD4 expression when compared to EGF group. FZD4 expression was at the highest level in the IGF-I group and this difference was statistically significant for all groups including uncultured cells and vehicle group. In addition, FGF-β was shown to positively affect the adhesion of granulosa cells. This study demonstrates that IGF-I, FGF-β and EGF have differential effects on the expressions of WNT2, WNT4, and FZD4 in cultured mouse granulosa cells, suggesting that particular growth factors related to ovarian function might conduct their roles in the ovary through Wnt signaling.
Collapse
Affiliation(s)
- Filiz Tepekoy
- Department of Histology and Embryology, Faculty of Medicine, Altinbas University, Istanbul, Turkey.,Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Gokhan Akkoyunlu
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
208
|
Ya J, Li X, Wang L, Kou H, Wang H, Zhao H. The effects of chronic cadmium exposure on the gut of Bufo gargarizans larvae at metamorphic climax: Histopathological impairments, microbiota changes and intestinal remodeling disruption. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 195:110523. [PMID: 32222598 DOI: 10.1016/j.ecoenv.2020.110523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 06/10/2023]
Abstract
Cadmium (Cd) is carcinogenic to human and it also has adverse effects on aquatic life such as amphibian larvae. However, its influences on amphibian gut morphology and development as well as intestinal microbiota are still hardly understood. In this study, we examined the effects of chronic cadmium exposure on the gut of tadpoles at Gosner stage 42 of metamorphic climax by using Bufo gargarizans as a model species. Tadpoles were exposed to cadmium concentrations at 0, 5, 100 and 200 μg L-1 from Gosner stage 26-42. The results showed that high cadmium (100 and 200 μg L-1) exposure caused significant decrease of body length and weight but significant increase of intestinal length and weight. Moreover, severe histopathological damages were induced by high Cd exposure. In addition, microbial communities in the gut of tadpoles in high cadmium exposure groups were remarkably different from those in control group. Unexpectedly, species diversity and richness were higher in the intestinal microbiota of 200 μg L-1 cadmium exposure group. Furthermore, the abundance of prevalent phyla, families and genera of intestinal microbiota were changed by cadmium exposure. Meanwhile, cadmium exposure perturbed gut renewal functions and the relative mRNA expression of genes involved in canonical and non-canonical Wnt signaling pathway was seriously affected by high cadmium exposure. We concluded that cadmium could be harmful to tadpole health by inducing intestinal histopathological damages, gut remodeling inhibition and intestinal microbiota alterations.
Collapse
Affiliation(s)
- Jing Ya
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Xuan Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Ling Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Honghong Kou
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Hongyuan Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Hongfeng Zhao
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China.
| |
Collapse
|
209
|
Holzem M, Franke FA, Mendes CC, McGregor AP. Wnt gene regulation and function during maxillary palp development in Drosophila melanogaster. Dev Biol 2020; 462:66-73. [PMID: 32229133 DOI: 10.1016/j.ydbio.2020.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 10/24/2022]
Abstract
Wnt genes encode secreted ligands that play many important roles in the development of metazoans. There are thirteen known Wnt gene subfamilies and seven of these are represented in Drosophila melanogaster. While wingless (wg) is the best understood and most widely studied Wnt gene in Drosophila, the functions of many of the other Drosophila Wnt genes are less well understood. For example, relatively little is known about Wnt6, which is an ancient paralog of wg and they form a conserved Wnt cluster together with Wnt9 (Dwnt4) and Wnt10. Wg and Wnt6 encode similar proteins and exhibit overlapping expression in several tissues during development. Both wg and Wnt6 were previously shown to regulate the development of maxillary palps, important olfactory organs in flies, but it remained unclear how these two ligands may combine to carry out specific functions and how this is regulated. Here, we have further analysed Wnt6 function in the context of maxillary palp development. Surprisingly, we found that Wnt6 does not appear to be necessary for development of maxillary palps. While a deletion of the 5' region of Wnt6 results in very small maxillary palps, we show that this effect is more likely to be a consequence of removing cis-regulatory elements that may regulate wg expression in this tissue rather than through the loss of Wnt6 function. Although, we cannot completely exclude the possibility that Wnt6 may subtly regulate maxillary palp development in combination with wg, our analysis of Wnt6 loss of function mutants suggests this ligand plays a more general role in regulating growth during development. Taken together our results provide new insights into maxillary palp formation and Wnt6 functions in Drosophila, and further evidence for a complex cis-regulatory landscape in the Wnt9-wg-Wnt6-Wnt10 cluster, which may help explain its evolutionary conservation.
Collapse
Affiliation(s)
- Michaela Holzem
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, OX3 0BP, United Kingdom; Division of Signalling and Functional Genomics, German Cancer Research Centre (DKFZ), Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
| | - Franziska A Franke
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, OX3 0BP, United Kingdom
| | - Cláudia C Mendes
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, OX3 0BP, United Kingdom; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX, United Kingdom
| | - Alistair P McGregor
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, OX3 0BP, United Kingdom.
| |
Collapse
|
210
|
Tran BM, Flanagan DJ, Ebert G, Warner N, Tran H, Fifis T, Kastrappis G, Christophi C, Pellegrini M, Torresi J, Phesse TJ, Vincan E. The Hepatitis B Virus Pre-Core Protein p22 Activates Wnt Signaling. Cancers (Basel) 2020; 12:cancers12061435. [PMID: 32486480 PMCID: PMC7352296 DOI: 10.3390/cancers12061435] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/24/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
An emerging theme for Wnt-addicted cancers is that the pathway is regulated at multiple steps via various mechanisms. Infection with hepatitis B virus (HBV) is a major risk factor for liver cancer, as is deregulated Wnt signaling, however, the interaction between these two causes is poorly understood. To investigate this interaction, we screened the effect of the various HBV proteins for their effect on Wnt/β-catenin signaling and identified the pre-core protein p22 as a novel and potent activator of TCF/β-catenin transcription. The effect of p22 on TCF/β-catenin transcription was dose dependent and inhibited by dominant-negative TCF4. HBV p22 activated synthetic and native Wnt target gene promoter reporters, and TCF/β-catenin target gene expression in vivo. Importantly, HBV p22 activated Wnt signaling on its own and in addition to Wnt or β-catenin induced Wnt signaling. Furthermore, HBV p22 elevated TCF/β-catenin transcription above constitutive activation in colon cancer cells due to mutations in downstream genes of the Wnt pathway, namely APC and CTNNB1. Collectively, our data identifies a previously unappreciated role for the HBV pre-core protein p22 in elevating Wnt signaling. Understanding the molecular mechanisms of p22 activity will provide insight into how Wnt signaling is fine-tuned in cancer.
Collapse
Affiliation(s)
- Bang Manh Tran
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne 3000, Australia; (B.M.T.); (D.J.F.)
| | - Dustin James Flanagan
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne 3000, Australia; (B.M.T.); (D.J.F.)
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Gregor Ebert
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (G.E.); (H.T.); (M.P.)
- Department of Medical Biology, The University of Melbourne, Melbourne 3010, Australia
| | - Nadia Warner
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Australia;
| | - Hoanh Tran
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (G.E.); (H.T.); (M.P.)
- Department of Medical Biology, The University of Melbourne, Melbourne 3010, Australia
| | - Theodora Fifis
- Department of Surgery, Austin Health, The University of Melbourne, Melbourne 3010, Australia; (T.F.); (G.K.); (C.C.)
| | - Georgios Kastrappis
- Department of Surgery, Austin Health, The University of Melbourne, Melbourne 3010, Australia; (T.F.); (G.K.); (C.C.)
| | - Christopher Christophi
- Department of Surgery, Austin Health, The University of Melbourne, Melbourne 3010, Australia; (T.F.); (G.K.); (C.C.)
| | - Marc Pellegrini
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (G.E.); (H.T.); (M.P.)
- Department of Medical Biology, The University of Melbourne, Melbourne 3010, Australia
| | - Joseph Torresi
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne 3000, Australia;
| | - Toby James Phesse
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne 3000, Australia; (B.M.T.); (D.J.F.)
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
- Correspondence: (T.J.P.); (E.V.); Tel.: +44-0-29-2068-849 (T.J.P.); +613 9342 9348 (E.V.)
| | - Elizabeth Vincan
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne 3000, Australia; (B.M.T.); (D.J.F.)
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Australia;
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia
- Correspondence: (T.J.P.); (E.V.); Tel.: +44-0-29-2068-849 (T.J.P.); +613 9342 9348 (E.V.)
| |
Collapse
|
211
|
Alvarez-Zavala M, Barreto-Vargas C, Torres-Reyes LA, De la Peña-Castro RF, Aguilar-Lemarroy A, Jave-Suarez LF. Exogenous Expression of WNT7A in Leukemia-Derived Cell Lines Induces Resistance to Chemotherapeutic Agents. Anticancer Agents Med Chem 2020; 20:1504-1514. [PMID: 32436833 DOI: 10.2174/1871520620666200521114100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/08/2020] [Accepted: 02/28/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dysregulations of the WNT pathway are implicated in the malignant transformation of different types of neoplasia. WNT7A is expressed in normal peripheral lymphocytes, but is decreased in the tumoral counterpart. Furthermore, the treatment of leukemic cells with recombinant WNT7A decreases proliferation, suggesting its possible use as a therapeutic biomolecule. This study aimed to evaluate the concomitant action of WNT7A and different chemotherapeutic agents over proliferation and cell death of leukemia/ lymphoma derived cell lines. METHODS Ectopic expression of WNT7A was induced in CEM and BJAB cell lines by using a lentiviral system. RNA expression was analyzed by microarrays and qPCR, and protein expression was determined by Western Blot. Cell proliferation was measured by cell counting, metabolic activity by WST-1 assay, cell death and DNA content by flow cytometry. RESULTS WNT7A ectopic expression was shown to decrease cell proliferation, but the apoptosis rate of leukemic cells was not altered. Moreover, these cells acquired resistance to doxorubicin, vincristine and MG-132. Cell cycle analysis reveals a decrease in G1 and an increase in S and G2 phases with a further upregulation of senescence- associated genes. Microarray analysis reveals that most gene expression changes were related to cancer and metabolic associated pathways. All those changes appear to be independent of the WNT canonical pathway regulation. CONCLUSION WNT7A negatively regulates cell proliferation in leukemic cell lines and promotes resistance to chemotherapeutic agents by inducing a senescence-like phenotype independently of the WNT canonical pathway.
Collapse
Affiliation(s)
- Monserrat Alvarez-Zavala
- Instituto de Investigacion en Inmunodeficiencias y VIH, Departamento de Clínicas Medicas, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Christian Barreto-Vargas
- Division de Inmunologia, Centro de Investigacion Biomedica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Luis A Torres-Reyes
- Departamento de Biologia Molecular y Genomica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Roberto F De la Peña-Castro
- Division de Inmunologia, Centro de Investigacion Biomedica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Adriana Aguilar-Lemarroy
- Division de Inmunologia, Centro de Investigacion Biomedica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Luis F Jave-Suarez
- Division de Inmunologia, Centro de Investigacion Biomedica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| |
Collapse
|
212
|
Cerbán FM, Stempin CC, Volpini X, Carrera Silva EA, Gea S, Motran CC. Signaling pathways that regulate Trypanosoma cruzi infection and immune response. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165707. [DOI: 10.1016/j.bbadis.2020.165707] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023]
|
213
|
Flores-Hernández E, Velázquez DM, Castañeda-Patlán MC, Fuentes-García G, Fonseca-Camarillo G, Yamamoto-Furusho JK, Romero-Avila MT, García-Sáinz JA, Robles-Flores M. Canonical and non-canonical Wnt signaling are simultaneously activated by Wnts in colon cancer cells. Cell Signal 2020; 72:109636. [PMID: 32283254 DOI: 10.1016/j.cellsig.2020.109636] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 12/26/2022]
Abstract
The Wnt signaling pathway is a crucial regulator of the intestinal epithelium homeostasis and is altered in most colon cancers. While the role of aberrant canonical, β-catenin-dependent Wnt signaling has been well established in colon cancer promotion, much less is known about the role played by noncanonical, β-catenin-independent Wnt signaling in this type of cancer. This work aimed to characterize the noncanonical signal transduction pathway in colon cancer cells. To this end, we used the prototype noncanonical ligand, Wnt5a, in comparison with Wnt3a, the prototype of a canonical β-catenin activating ligand. The analysis of the expression profile of Wnt receptors in colon cancer cell lines showed a clear increase in both level expression and variety of Frizzled receptor types expressed in colon cancer cells compared with non-malignant cells. We found that Wnt5a activates a typical Wnt/Ca++ - noncanonical signaling pathway in colon malignant cells, inducing the hyperphosphorylation of Dvl1, Dvl2 and Dvl3, promoting Ca++ mobilization as a result of phospholipase C (PLC) activation via pertussis toxin-sensitive G-protein, and inducing PLC-dependent cell migration. We also found that while the co-receptor Ror2 tyrosine kinase activity is not required for Ca++ mobilization-induced by Wnt5a, it is required for the inhibitory effects of Wnt5a on the β-catenin-dependent transcriptional activity. Unexpectedly, we found that although the prototype canonical Wnt3a ligand was unique in stimulating the β-catenin-dependent transcriptional activity, it also simultaneously activated PLC, promoted Ca++ mobilization, and induced Rho kinase and PLC-dependent cell migration. Our data indicate, therefore, that a Wnt ligand can activate at the same time the so-called Wnt canonical and noncanonical pathways inducing the formation of complex signaling networks to integrate both pathways in colon cancer cells.
Collapse
Affiliation(s)
- Eric Flores-Hernández
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Dora M Velázquez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - M Cristina Castañeda-Patlán
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Gabriela Fuentes-García
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Gabriela Fonseca-Camarillo
- Inflammatory Bowel Disease Clinic, Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jesús K Yamamoto-Furusho
- Inflammatory Bowel Disease Clinic, Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - M Teresa Romero-Avila
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - J Adolfo García-Sáinz
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Martha Robles-Flores
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.
| |
Collapse
|
214
|
Sidrat T, Khan AA, Idrees M, Joo MD, Xu L, Lee KL, Kong IK. Role of Wnt Signaling During In-Vitro Bovine Blastocyst Development and Maturation in Synergism with PPARδ Signaling. Cells 2020; 9:cells9040923. [PMID: 32283810 PMCID: PMC7226827 DOI: 10.3390/cells9040923] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
Wnt/β-catenin signaling plays vital role in the regulation of cellular proliferation, migration, stem cells cell renewal and genetic stability. This pathway is crucial during the early developmental process; however, the distinct role of Wnt/β-catenin signaling during pre-implantation period of bovine embryonic development is obscure. Here, we evaluated the critical role of Wnt/β-catenin pathway in the regulation of bovine blastocyst (BL) development and hatching. 6 bromoindurbin-3’oxime (6-Bio) was used to stimulate the Wnt signaling. Treatment with 6-Bio induced the expression of peroxisome proliferator-activated receptor-delta (PPARδ). Interestingly, the PPARδ co-localized with β-catenin and form a complex with TCF/LEF transcription factor. This complex potentiated the expression of several Wnt directed genes, which regulate early embryonic development. Inhibition of PPARδ with selective inhibitor 4-chloro-N-(2-{[5-trifluoromethyl]-2-pyridyl]sulfonyl}ethyl)benzamide (Gsk3787) severely perturbed the BL formation and hatching. The addition of Wnt agonist successfully rescued the BL formation and hatching ability. Importantly, the activation of PPARδ expression by Wnt stimulation enhanced cell proliferation and fatty acid oxidation (FAO) metabolism to improve BL development and hatching. In conclusion, our study provides the evidence that Wnt induced PPARδ expression co-localizes with β-catenin and is a likely candidate of canonical Wnt pathway for the regulation of bovine embryonic development.
Collapse
Affiliation(s)
- Tabinda Sidrat
- Department of Animal Science, Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (T.S.); (M.I.); (L.X.)
| | - Abdul Aziz Khan
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA;
| | - Muhammad Idrees
- Department of Animal Science, Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (T.S.); (M.I.); (L.X.)
| | - Myeong-Don Joo
- Department of Animal Science, Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (T.S.); (M.I.); (L.X.)
| | - Lianguang Xu
- Department of Animal Science, Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (T.S.); (M.I.); (L.X.)
| | - Kyeong-Lim Lee
- Department of Animal Science, Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (T.S.); (M.I.); (L.X.)
| | - Il-Keun Kong
- Department of Animal Science, Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (T.S.); (M.I.); (L.X.)
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Korea
- Correspondence: ; Tel.: +82-55-772-1942
| |
Collapse
|
215
|
Leng S, Pignatti E, Khetani RS, Shah MS, Xu S, Miao J, Taketo MM, Beuschlein F, Barrett PQ, Carlone DL, Breault DT. β-Catenin and FGFR2 regulate postnatal rosette-based adrenocortical morphogenesis. Nat Commun 2020; 11:1680. [PMID: 32245949 PMCID: PMC7125176 DOI: 10.1038/s41467-020-15332-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 02/28/2020] [Indexed: 02/08/2023] Open
Abstract
Rosettes are widely used in epithelial morphogenesis during embryonic development and organogenesis. However, their role in postnatal development and adult tissue maintenance remains largely unknown. Here, we show zona glomerulosa cells in the adult adrenal cortex organize into rosettes through adherens junction-mediated constriction, and that rosette formation underlies the maturation of adrenal glomerular structure postnatally. Using genetic mouse models, we show loss of β-catenin results in disrupted adherens junctions, reduced rosette number, and dysmorphic glomeruli, whereas β-catenin stabilization leads to increased adherens junction abundance, more rosettes, and glomerular expansion. Furthermore, we uncover numerous known regulators of epithelial morphogenesis enriched in β-catenin-stabilized adrenals. Among these genes, we show Fgfr2 is required for adrenal rosette formation by regulating adherens junction abundance and aggregation. Together, our data provide an example of rosette-mediated postnatal tissue morphogenesis and a framework for studying the role of rosettes in adult zona glomerulosa tissue maintenance and function.
Collapse
Affiliation(s)
- Sining Leng
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Division of Medical Sciences, Harvard Medical School, Boston, MA, 02115, USA
| | - Emanuele Pignatti
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Radhika S Khetani
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Manasvi S Shah
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Simiao Xu
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Makoto M Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-Cho, Sakyo, Kyoto, 606-8506, Japan
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, UniversitätsSpital Zürich, Zurich, Switzerland
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Paula Q Barrett
- Departments of Pharmacology, University of Virginia, Charlottesville, VA, 22947, USA
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
216
|
Arisan ED, Rencuzogullari O, Freitas IL, Radzali S, Keskin B, Kothari A, Warford A, Uysal-Onganer P. Upregulated Wnt-11 and miR-21 Expression Trigger Epithelial Mesenchymal Transition in Aggressive Prostate Cancer Cells. BIOLOGY 2020; 9:biology9030052. [PMID: 32182839 PMCID: PMC7150874 DOI: 10.3390/biology9030052] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/24/2020] [Accepted: 03/06/2020] [Indexed: 01/31/2023]
Abstract
Prostate cancer (PCa) is the second-leading cause of cancer-related death among men. microRNAs have been identified as having potential roles in tumorigenesis. An oncomir, miR-21, is commonly highly upregulated in many cancers, including PCa, and showed correlation with the Wnt-signaling axis to increase invasion. Wnt-11 is a developmentally regulated gene and has been found to be upregulated in PCa, but its mechanism is unknown. The present study aimed to investigate the roles of miR-21 and Wnt-11 in PCa in vivo and in vitro. First, different Gleason score PCa tissue samples were used; both miR-21 and Wnt-11 expressions correlate with high Gleason scores in PCa patient tissues. This data then was confirmed with formalin-fixed paraffin cell blocks using PCa cell lines LNCaP and PC3. Cell survival and colony formation studies proved that miR-21 involves in cells’ behaviors, as well as the epithelial-mesenchymal transition. Consistent with the previous data, silencing miR-21 led to significant inhibition of cellular invasiveness. Overall, these results suggest that miR-21 plays a significant role related to Wnt-11 in the pathophysiology of PCa.
Collapse
Affiliation(s)
- Elif Damla Arisan
- Institute of Biotechnology, Gebze Technical University, Gebze 41400, Kocaeli, Turkey;
| | - Ozge Rencuzogullari
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Atakoy Campus 34156, Istanbul, Turkey; (O.R.); (B.K.)
| | - Ines Lua Freitas
- Cancer Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (I.L.F.); ; (S.R.); (A.W.)
| | - Syanas Radzali
- Cancer Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (I.L.F.); ; (S.R.); (A.W.)
| | - Buse Keskin
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Atakoy Campus 34156, Istanbul, Turkey; (O.R.); (B.K.)
| | - Archana Kothari
- Department of Histopathology, Kingston Hospital, Galsworthy Road, London KT2 7QE, UK;
| | - Antony Warford
- Cancer Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (I.L.F.); ; (S.R.); (A.W.)
| | - Pinar Uysal-Onganer
- Cancer Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (I.L.F.); ; (S.R.); (A.W.)
- Correspondence: ; Tel.: +44-(0)207-911-5151 (ext. 64581)
| |
Collapse
|
217
|
Lobo IKC, Nascimento ÁRD, Yamagishi MEB, Guiguen Y, Silva GFD, Severac D, Amaral ADC, Reis VR, Almeida FLD. Transcriptome of tambaqui Colossoma macropomum during gonad differentiation: Different molecular signals leading to sex identity. Genomics 2020; 112:2478-2488. [PMID: 32027957 DOI: 10.1016/j.ygeno.2020.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 01/11/2020] [Accepted: 01/31/2020] [Indexed: 12/13/2022]
Abstract
Tambaqui (Colossoma macropomum) is the major native species in Brazilian aquaculture, and we have shown that females exhibit a higher growth compared to males, opening up the possibility for the production of all-female population. To date, there is no information on the sex determination and differentiation molecular mechanisms of tambaqui. In the present study, transcriptome sequencing of juvenile trunks was performed to understand the molecular network involved in the gonadal sex differentiation. The results showed that before differentiation, components of the Wnt/β-catenin pathway, fox and fst genes imprint female sex development, whereas antagonistic pathways (gsk3b, wt1 and fgfr2), sox9 and genes for androgen synthesis indicate male differentiation. Hence, in undifferentiated tambaqui, the Wnt/β-catenin exerts a role on sex differentiation, either upregulated in female-like individuals, or antagonized in male-like individuals.
Collapse
Affiliation(s)
| | | | | | - Yann Guiguen
- INRA, UR1037 LPGP, Campus de Beaulieu, Rennes, France.
| | | | - Dany Severac
- MGX, Univ Montpellier, CNRS, INSERM, Montpellier, France.
| | - Aldessandro da Costa Amaral
- Programa de Pós-graduação em Ciências Pesqueiras nos Trópicos, Universidade Federal do Amazonas, Manaus, Brazil
| | - Vanessa Ribeiro Reis
- Programa de Pós-graduação em Biotecnologia, Universidade Federal do Amazonas, Manaus, Brazil
| | | |
Collapse
|
218
|
van Schie EH, van Amerongen R. Aberrant WNT/CTNNB1 Signaling as a Therapeutic Target in Human Breast Cancer: Weighing the Evidence. Front Cell Dev Biol 2020; 8:25. [PMID: 32083079 PMCID: PMC7005411 DOI: 10.3389/fcell.2020.00025] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022] Open
Abstract
WNT signaling is crucial for tissue morphogenesis during development in all multicellular animals. After birth, WNT/CTNNB1 responsive stem cells are responsible for tissue homeostasis in various organs and hyperactive WNT/CTNNB1 signaling is observed in many different human cancers. The first link between WNT signaling and breast cancer was established almost 40 years ago, when Wnt1 was identified as a proto-oncogene capable of driving mammary tumor formation in mice. Since that discovery, there has been a dedicated search for aberrant WNT signaling in human breast cancer. However, much debate and controversy persist regarding the importance of WNT signaling for the initiation, progression or maintenance of different breast cancer subtypes. As the first drugs designed to block functional WNT signaling have entered clinical trials, many questions about the role of aberrant WNT signaling in human breast cancer remain. Here, we discuss three major research gaps in this area. First, we still lack a basic understanding of the function of WNT signaling in normal human breast development and physiology. Second, the overall extent and precise effect of (epi)genetic changes affecting the WNT pathway in different breast cancer subtypes are still unknown. Which underlying molecular and cell biological mechanisms are disrupted as a result also awaits further scrutiny. Third, we survey the current status of targeted therapeutics that are aimed at interfering with the WNT pathway in breast cancer patients and highlight the importance and complexity of selecting the subset of patients that may benefit from treatment.
Collapse
Affiliation(s)
| | - Renée van Amerongen
- Section of Molecular Cytology and van Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
219
|
Hsu WL, Ma YL, Liu YC, Tai DJC, Lee EHY. Restoring Wnt6 signaling ameliorates behavioral deficits in MeCP2 T158A mouse model of Rett syndrome. Sci Rep 2020; 10:1074. [PMID: 31974426 PMCID: PMC6978308 DOI: 10.1038/s41598-020-57745-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/09/2019] [Indexed: 01/23/2023] Open
Abstract
The methyl-CpG-binding protein 2 gene, MECP2, is an X chromosome-linked gene encoding the MeCP2 protein, and mutations of MECP2 cause Rett syndrome (RTT). Previous study has shown that re-expression of SUMO-modified MeCP2 in Mecp2-null neurons rescues synaptic and behavioral deficits in Mecp2 conditional knockout mice, whereas about 12-fold decrease in Wnt6 mRNA level was found in MeCP2K412R sumo-mutant mice. Here, we examined the role of Wnt6 in MeCP2 T158A mouse model of RTT. Results show that lentiviral delivery of Wnt6 to the amygdala ameliorates locomotor impairment and social behavioral deficits in these animals. MeCP2 T158A mice show decreased level of GSK-3β phosphorylation and increased level of β-catenin phosphorylation. They also show reduced level of MeCP2 SUMOylation. These alterations were also restored by lenti-Wnt6 transduction. Further, both BDNF and IGF-1 expressions are decreased in MeCP2 T158A mice. Overexpression of Wnt6 increases Bdnf and Igf-1 promoter activity in HEK293T cells in a dose-dependent manner. Lenti-Wnt6 transduction to the amygdala similarly increases the mRNA level and protein expression of BDNF and IGF-1 in MeCP2 T158A mice. Moreover, environmental enrichment (EE) similarly ameliorates the locomotor and social behavioral deficits in MeCP2 T158A mice. One of the mechanisms underlying EE is mediated through enhanced MeCP2 SUMOylation and increased Wnt6 expression in these animals by EE.
Collapse
Affiliation(s)
- Wei-Lun Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yun-Li Ma
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yen-Chen Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Derek J C Tai
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Eminy H Y Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
220
|
Levin G, Koga BAA, Belchior GG, Carreira ACO, Sogayar MC. Production, purification and characterization of recombinant human R-spondin1 (RSPO1) protein stably expressed in human HEK293 cells. BMC Biotechnol 2020; 20:5. [PMID: 31959207 PMCID: PMC6971977 DOI: 10.1186/s12896-020-0600-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 01/13/2020] [Indexed: 02/07/2023] Open
Abstract
Background The R-Spondin proteins comprise a family of secreted proteins, known for their important roles in cell proliferation, differentiation and death, by inducing the Wnt pathway. Several studies have demonstrated the importance of RSPOs in regulation of a number of tissue-specific processes, namely: bone formation, skeletal muscle tissue development, proliferation of pancreatic β-cells and intestinal stem cells and even cancer. RSPO1 stands out among RSPOs molecules with respect to its potential therapeutic use, especially in the Regenerative Medicine field, due to its mitogenic activity in stem cells. Here, we generated a recombinant human RSPO1 (rhRSPO1) using the HEK293 cell line, obtaining a purified, characterized and biologically active protein product to be used in Cell Therapy. The hRSPO1 coding sequence was synthesized and subcloned into a mammalian cell expression vector. HEK293 cells were stably co-transfected with the recombinant expression vector containing the hRSPO1 coding sequence and a hygromycin resistance plasmid, selected for hygror and subjected to cell clones isolation. Results rhRSPO1 was obtained, in the absence of serum, from culture supernatants of transfected HEK293 cells and purified using a novel purification strategy, involving two sequential chromatographic steps, namely: heparin affinity chromatography, followed by a molecular exclusion chromatography, designed to yield a high purity product. The purified protein was characterized by Western blotting, mass spectrometry and in vitro (C2C12 cells) and in vivo (BALB/c mice) biological activity assays, confirming the structural integrity and biological efficacy of this human cell expression system. Furthermore, rhRSPO1 glycosylation analysis allowed us to describe, for the first time, the glycan composition of this oligosaccharide chain, confirming the presence of an N-glycosylation in residue Asn137 of the polypeptide chain, as previously described. In addition, this analysis revealing the presence of glycan structures such as terminal sialic acid, N-acetylglucosamine and/or galactose. Conclusion Therefore, a stable platform for the production and purification of recombinant hRSPO1 from HEK293 cells was generated, leading to the production of a purified, fully characterized and biologically active protein product to be applied in Tissue Engineering.
Collapse
Affiliation(s)
- Gabriel Levin
- Cell and Molecular Therapy Center (NUCEL), Medical School, University of São Paulo, Edifício NUCEL, Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil
| | - Bruna Andrade Aguiar Koga
- Cell and Molecular Therapy Center (NUCEL), Medical School, University of São Paulo, Edifício NUCEL, Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil.,Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, SP, 13635-900, Brazil
| | - Gustavo Gross Belchior
- Cell and Molecular Therapy Center (NUCEL), Medical School, University of São Paulo, Edifício NUCEL, Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil
| | - Ana Claudia Oliveira Carreira
- Cell and Molecular Therapy Center (NUCEL), Medical School, University of São Paulo, Edifício NUCEL, Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil. .,Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, SP, 13635-900, Brazil.
| | - Mari Cleide Sogayar
- Cell and Molecular Therapy Center (NUCEL), Medical School, University of São Paulo, Edifício NUCEL, Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil. .,Biochemistry Department, Chemistry Institute, University of São Paulo, Sao Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
221
|
Ashrafizadeh M, Ahmadi Z, Mohamamdinejad R, Yaribeygi H, Serban MC, Orafai HM, Sahebkar A. Curcumin Therapeutic Modulation of the Wnt Signaling Pathway. Curr Pharm Biotechnol 2020; 21:1006-1015. [PMID: 32133961 DOI: 10.2174/1389201021666200305115101] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/20/2019] [Accepted: 01/18/2020] [Indexed: 01/21/2023]
Abstract
Curcumin, isolated from the rhizome of Curcuma longa, is one of the most extensively studied phytochemicals. This natural compound has a variety of pharmacological effects including antioxidant, anti-inflammatory, anti-tumor, cardio-protective, hepato-protective and anti-diabetic. Wnt signaling pathway, one of the potential targets of curcumin through upregulation and/or downregulation, plays a significant role in many diseases, even in embryogenesis and development of various organs and systems. In order to exert an anti-tumor activity in the organism, curcumin seems to inhibit the Wnt pathway. The downstream mediators of Wnt signaling pathway such as c-Myc and cyclin D1 are also modified by curcumin. This review demonstrates how curcumin influences the Wnt signaling pathway and is beneficial for the treatment of neurological disorders (Alzheimer's and Parkinson's diseases), cancers (melanoma, lung cancer, breast cancer, colon cancer, endothelial carcinoma, gastric carcinoma and hepatocellular carcinoma) and other diseases, such as diabetes mellitus or bone disorders.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Zahra Ahmadi
- Department of Basic Science, Veterinary Medicine Faculty, Shushtar University, Khuzestan, Iran
| | - Reza Mohamamdinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Romania
| | - Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Maria-Corina Serban
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Hossein M Orafai
- Department of Pharmaceutics, Faculty of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
| | | |
Collapse
|
222
|
Huybrechts Y, Mortier G, Boudin E, Van Hul W. WNT Signaling and Bone: Lessons From Skeletal Dysplasias and Disorders. Front Endocrinol (Lausanne) 2020; 11:165. [PMID: 32328030 PMCID: PMC7160326 DOI: 10.3389/fendo.2020.00165] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Skeletal dysplasias are a diverse group of heritable diseases affecting bone and cartilage growth. Throughout the years, the molecular defect underlying many of the diseases has been identified. These identifications led to novel insights in the mechanisms regulating bone and cartilage growth and homeostasis. One of the pathways that is clearly important during skeletal development and bone homeostasis is the Wingless and int-1 (WNT) signaling pathway. So far, three different WNT signaling pathways have been described, which are all activated by binding of the WNT ligands to the Frizzled (FZD) receptors. In this review, we discuss the skeletal disorders that are included in the latest nosology of skeletal disorders and that are caused by genetic defects involving the WNT signaling pathway. The number of skeletal disorders caused by defects in WNT signaling genes and the clinical phenotype associated with these disorders illustrate the importance of the WNT signaling pathway during skeletal development as well as later on in life to maintain bone mass. The knowledge gained through the identification of the genes underlying these monogenic conditions is used for the identification of novel therapeutic targets. For example, the genes underlying disorders with altered bone mass are all involved in the canonical WNT signaling pathway. Consequently, targeting this pathway is one of the major strategies to increase bone mass in patients with osteoporosis. In addition to increasing the insights in the pathways regulating skeletal development and bone homeostasis, knowledge of rare skeletal dysplasias can also be used to predict possible adverse effects of these novel drug targets. Therefore, this review gives an overview of the skeletal and extra-skeletal phenotype of the different skeletal disorders linked to the WNT signaling pathway.
Collapse
|
223
|
Cardoso BA. The Bone Marrow Niche - The Tumor Microenvironment That Ensures Leukemia Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:259-293. [PMID: 32130704 DOI: 10.1007/978-3-030-34025-4_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The human body requires a constant delivery of fresh blood cells that are needed to maintain body homeostasis. Hematopoiesis is the process that drives the formation of new blood cells from a single stem cell. This is a complex, orchestrated and tightly regulated process that occurs within the bone marrow. When such process is faulty or deregulated, leukemia arises, develops and thrives by subverting normal hematopoiesis and availing the supplies of this rich milieu.In this book chapter we will describe and characterize the bone marrow microenvironment and its key importance for leukemia expansion. The several components of the bone marrow niche, their interaction with the leukemic cells and the cellular pathways activated within the malignant cells will be emphasized. Finally, novel therapeutic strategies to target this sibling interaction will also be discussed.
Collapse
Affiliation(s)
- Bruno António Cardoso
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
224
|
Martínez-Bartolomé M, Range RC. A biphasic role of non-canonical Wnt16 signaling during early anterior-posterior patterning and morphogenesis of the sea urchin embryo. Development 2019; 146:dev168799. [PMID: 31822478 PMCID: PMC6955209 DOI: 10.1242/dev.168799] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 11/26/2019] [Indexed: 12/28/2022]
Abstract
A Wnt signaling network governs early anterior-posterior (AP) specification and patterning of the deuterostome sea urchin embryo. We have previously shown that non-canonical Fzl1/2/7 signaling antagonizes the progressive posterior-to-anterior downregulation of the anterior neuroectoderm (ANE) gene regulatory network (GRN) by canonical Wnt/β-catenin and non-canonical Wnt1/Wnt8-Fzl5/8-JNK signaling. This study focuses on the non-canonical function of the Wnt16 ligand during early AP specification and patterning. Maternally supplied wnt16 is expressed ubiquitously during cleavage and zygotic wnt16 expression is concentrated in the endoderm/mesoderm beginning at mid-blastula stage. Wnt16 antagonizes the ANE restriction mechanism and this activity depends on a functional Fzl1/2/7 receptor. Our results also show that zygotic wnt16 expression depends on both Fzl5/8 and Wnt/β-catenin signaling. Furthermore, Wnt16 is necessary for the activation and/or maintenance of key regulatory endoderm/mesoderm genes and is essential for gastrulation. Together, our data show that Wnt16 has two functions during early AP specification and patterning: (1) an initial role activating the Fzl1/2/7 pathway that antagonizes the ANE restriction mechanism; and (2) a subsequent function in activating key endoderm GRN factors and the morphogenetic movements of gastrulation.
Collapse
Affiliation(s)
| | - Ryan C Range
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
225
|
Patel S, Alam A, Pant R, Chattopadhyay S. Wnt Signaling and Its Significance Within the Tumor Microenvironment: Novel Therapeutic Insights. Front Immunol 2019; 10:2872. [PMID: 31921137 PMCID: PMC6927425 DOI: 10.3389/fimmu.2019.02872] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 11/22/2019] [Indexed: 01/05/2023] Open
Abstract
Wnt signaling is one of the central mechanisms regulating tissue morphogenesis during embryogenesis and repair. The pivot of this signaling cascade is the Wnt ligand, which binds to receptors belonging to the Frizzled family or the ROR1/ROR2 and RYK family. This interaction governs the downstream signaling cascade (canonical/non-canonical), ultimately extending its effect on the cellular cytoskeleton, transcriptional control of proliferation and differentiation, and organelle dynamics. Anomalous Wnt signaling has been associated with several cancers, the most prominent ones being colorectal, breast, lung, oral, cervical, and hematopoietic malignancies. It extends its effect on tumorigenesis by modulating the tumor microenvironment via fine crosstalk between transformed cells and infiltrating immune cells, such as leukocytes. This review is an attempt to highlight the latest developments in the understanding of Wnt signaling in the context of tumors and their microenvironment. A dynamic process known as immunoediting governs the fate of tumor progression based on the correlation of various signaling pathways in the tumor microenvironment and immune cells. Cancer cells also undergo a series of mutations in the tumor suppressor gene, which favors tumorigenesis. Wnt signaling, and its crosstalk with various immune cells, has both negative as well as positive effects on tumor progression. On one hand, it helps in the maintenance and renewal of the leucocytes. On the other hand, it promotes immune tolerance, limiting the antitumor response. Wnt signaling also plays a role in epithelial-mesenchymal transition (EMT), thereby promoting the maintenance of Cancer Stem Cells (CSCs). Furthermore, we have summarized the ongoing strategies used to target aberrant Wnt signaling as a novel therapeutic intervention to combat various cancers and their limitations.
Collapse
Affiliation(s)
- Sonal Patel
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, India
| | - Aftab Alam
- Department of Cancer Biology and Inflammatory Disorder, Indian Institute of Chemical Biology, Kolkata, India
| | - Richa Pant
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, India
| | - Samit Chattopadhyay
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, India.,Department of Cancer Biology and Inflammatory Disorder, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
226
|
de Souza LC, Cavalla F, Maili L, Garlet GP, Vieira AR, Silva RM, Letra A. WNT gene polymorphisms and predisposition to apical periodontitis. Sci Rep 2019; 9:18980. [PMID: 31831777 PMCID: PMC6908593 DOI: 10.1038/s41598-019-55293-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Single nucleotide polymorphisms (SNPs) in WNT genes may impact gene/protein function and contribute to individual predisposition to apical periodontitis (AP). Here, we investigated the association of SNPs in/nearby WNT3, WNT3A, WNT5A, WNT8A, WNT9B and WNT11 genes with AP using a case-control dataset. Cases were defined as individuals with deep caries and AP (n = 188); controls had deep caries and no AP (n = 230). Genotyping was performed using Taqman chemistry in real time PCR. Data analyses was performed using Fisher Exact tests assuming a Bonferroni correction threshold value of 0.005. Single-SNP association analysis revealed a trend for association with WNT3 rs9890413 genotypes (P = 0.009) under a dominant model and allelic association for WNT3A rs1745420 (P = 0.009). Haplotypes involving WNT3-WNT9B-WNT3A alleles were also significantly associated with AP (P ≤ 0.003). Luciferase reporter assays showed higher transcriptional activity (1.4-fold) with the alternate G allele in rs1745420. Expression of WNT3, WNT3A and WNT5A in AP tissues was significantly higher than in control tissues, and inversely correlated with the expression of SERPINB1, COL1A1 and TIMP1 (P < 0.05). Our results suggest that WNT genes have a role in modulating AP and polymorphisms in these genes may increase susceptibility to AP.
Collapse
Affiliation(s)
- Letícia Chaves de Souza
- Department of Endodontics, University of Texas Health Science Center School of Dentistry, Houston, 77054, USA
- Center for Craniofacial Research, University of Texas Health Science Center School of Dentistry, Houston, 77054, USA
| | - Franco Cavalla
- Center for Craniofacial Research, University of Texas Health Science Center School of Dentistry, Houston, 77054, USA
- Department of Conservative Dentistry, School of Dentistry, University of Chile, Santiago, 7520355, Chile
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, 17012, Brazil
| | - Lorena Maili
- Department of Pediatrics, University of Texas Health Science Center at Houston McGovern Medical School, Houston, 77030, USA
- Pediatric Research Center, University of Texas Health Science Center at Houston McGovern Medical School, Houston, 77030, USA
| | - Gustavo P Garlet
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, 17012, Brazil
| | - Alexandre R Vieira
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, 15229, USA
| | - Renato M Silva
- Department of Endodontics, University of Texas Health Science Center School of Dentistry, Houston, 77054, USA
- Center for Craniofacial Research, University of Texas Health Science Center School of Dentistry, Houston, 77054, USA
| | - Ariadne Letra
- Center for Craniofacial Research, University of Texas Health Science Center School of Dentistry, Houston, 77054, USA.
- Pediatric Research Center, University of Texas Health Science Center at Houston McGovern Medical School, Houston, 77030, USA.
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center School of Dentistry, Houston, 77054, USA.
| |
Collapse
|
227
|
Young RM, Ewan KB, Ferrer VP, Allende ML, Godovac-Zimmermann J, Dale TC, Wilson SW. Developmentally regulated Tcf7l2 splice variants mediate transcriptional repressor functions during eye formation. eLife 2019; 8:e51447. [PMID: 31829936 PMCID: PMC6908431 DOI: 10.7554/elife.51447] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/24/2019] [Indexed: 12/11/2022] Open
Abstract
Tcf7l2 mediates Wnt/β-Catenin signalling during development and is implicated in cancer and type-2 diabetes. The mechanisms by which Tcf7l2 and Wnt/β-Catenin signalling elicit such a diversity of biological outcomes are poorly understood. Here, we study the function of zebrafish tcf7l2alternative splice variants and show that only variants that include exon five or an analogous human tcf7l2 variant can effectively provide compensatory repressor function to restore eye formation in embryos lacking tcf7l1a/tcf7l1b function. Knockdown of exon five specific tcf7l2 variants in tcf7l1a mutants also compromises eye formation, and these variants can effectively repress Wnt pathway activity in reporter assays using Wnt target gene promoters. We show that the repressive activities of exon5-coded variants are likely explained by their interaction with Tle co-repressors. Furthermore, phosphorylated residues in Tcf7l2 coded exon5 facilitate repressor activity. Our studies suggest that developmentally regulated splicing of tcf7l2 can influence the transcriptional output of the Wnt pathway.
Collapse
Affiliation(s)
- Rodrigo M Young
- Department of Cell and Developmental BiologyUCLLondonUnited Kingdom
| | - Kenneth B Ewan
- School of Bioscience, Cardiff UniversityCardiffUnited Kingdom
| | | | - Miguel L Allende
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de ChileSantiagoChile
| | | | - Trevor C Dale
- School of Bioscience, Cardiff UniversityCardiffUnited Kingdom
| | - Stephen W Wilson
- Department of Cell and Developmental BiologyUCLLondonUnited Kingdom
| |
Collapse
|
228
|
Predes D, Oliveira LFS, Ferreira LSS, Maia LA, Delou JMA, Faletti A, Oliveira I, Amado NG, Reis AH, Fraga CAM, Kuster R, Mendes FA, Borges HL, Abreu JG. The Chalcone Lonchocarpin Inhibits Wnt/β-Catenin Signaling and Suppresses Colorectal Cancer Proliferation. Cancers (Basel) 2019; 11:cancers11121968. [PMID: 31817828 PMCID: PMC6966512 DOI: 10.3390/cancers11121968] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 12/19/2022] Open
Abstract
The deregulation of the Wnt/β-catenin signaling pathway is a central event in colorectal cancer progression, thus a promising target for drug development. Many natural compounds, such as flavonoids, have been described as Wnt/β-catenin inhibitors and consequently modulate important biological processes like inflammation, redox balance, cancer promotion and progress, as well as cancer cell death. In this context, we identified the chalcone lonchocarpin isolated from Lonchocarpus sericeus as a Wnt/β-catenin pathway inhibitor, both in vitro and in vivo. Lonchocarpin impairs β-catenin nuclear localization and also inhibits the constitutively active form of TCF4, dnTCF4-VP16. Xenopus laevis embryology assays suggest that lonchocarpin acts at the transcriptional level. Additionally, we described lonchocarpin inhibitory effects on cell migration and cell proliferation on HCT116, SW480, and DLD-1 colorectal cancer cell lines, without any detectable effects on the non-tumoral intestinal cell line IEC-6. Moreover, lonchocarpin reduces tumor proliferation on the colorectal cancer AOM/DSS mice model. Taken together, our results support lonchocarpin as a novel Wnt/β-catenin inhibitor compound that impairs colorectal cancer cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Danilo Predes
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Luiz F. S. Oliveira
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Laís S. S. Ferreira
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Lorena A. Maia
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - João M. A. Delou
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Anderson Faletti
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Igor Oliveira
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Nathalia G. Amado
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Alice H. Reis
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Carlos A. M. Fraga
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ricardo Kuster
- Department of Chemistry, Federal University of Espírito Santo, Espírito Santo 29075-910, Brazil
| | - Fabio A. Mendes
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Helena L. Borges
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Jose G. Abreu
- Program of Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Correspondence: ; Tel.: +55-21-3938-6486
| |
Collapse
|
229
|
Lu M, Mizumoto K. Gradient-independent Wnt signaling instructs asymmetric neurite pruning in C. elegans. eLife 2019; 8:e50583. [PMID: 31804181 PMCID: PMC6894928 DOI: 10.7554/elife.50583] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022] Open
Abstract
During development, the nervous system undergoes a refinement process by which neurons initially extend an excess number of neurites, the majority of which will be eliminated by the mechanism called neurite pruning. Some neurites undergo stereotyped and developmentally regulated pruning. However, the signaling cues that instruct stereotyped neurite pruning are yet to be fully elucidated. Here we show that Wnt morphogen instructs stereotyped neurite pruning for proper neurite projection patterning of the cholinergic motor neuron called PDB in C. elegans. In lin-44/wnt and lin-17/frizzled mutant animals, the PDB neurites often failed to prune and grew towards the lin-44-expressing cells. Surprisingly, membrane-tethered lin-44 is sufficient to induce proper neurite pruning in PDB, suggesting that neurite pruning does not require a Wnt gradient. LIN-17 and DSH-1/Dishevelled proteins were recruited to the pruning neurites in lin-44-dependent manners. Our results revealed the novel gradient-independent role of Wnt signaling in instructing neurite pruning.
Collapse
Affiliation(s)
- Menghao Lu
- Department of ZoologyUniversity of British ColumbiaVancouverCanada
| | - Kota Mizumoto
- Department of ZoologyUniversity of British ColumbiaVancouverCanada
- Life Sciences Institute, University of British ColumbiaVancouverCanada
| |
Collapse
|
230
|
Zeng S, Ulbrich SE, Bauersachs S. Spatial organization of endometrial gene expression at the onset of embryo attachment in pigs. BMC Genomics 2019; 20:895. [PMID: 31752681 PMCID: PMC6873571 DOI: 10.1186/s12864-019-6264-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND During the preimplantation phase in the pig, the conceptus trophoblast elongates into a filamentous form and secretes estrogens, interleukin 1 beta 2, interferons, and other signaling molecules before attaching to the uterine epithelium. The processes in the uterine endometrium in response to conceptus signaling are complex. Thus, the objective of this study was to characterize transcriptome changes in porcine endometrium during the time of conceptus attachment considering the specific localization in different endometrial cell types. RESULTS Low-input RNA-sequencing was conducted for the main endometrial compartments, luminal epithelium (LE), glandular epithelium (GE), blood vessels (BV), and stroma. Samples were isolated from endometria collected on Day 14 of pregnancy and the estrous cycle (each group n = 4) by laser capture microdissection. The expression of 12,000, 11,903, 11,094, and 11,933 genes was detectable in LE, GE, BV, and stroma, respectively. Differential expression analysis was performed between the pregnant and cyclic group for each cell type as well as for a corresponding dataset for complete endometrium tissue samples. The highest number of differentially expressed genes (DEGs) was found for LE (1410) compared to GE, BV, and stroma (800, 1216, and 384). For the complete tissue, 3262 DEGs were obtained. The DEGs were assigned to Gene Ontology (GO) terms to find overrepresented functional categories and pathways specific for the individual endometrial compartments. GO classification revealed that DEGs in LE were involved in 'biosynthetic processes', 'related to ion transport', and 'apoptotic processes', whereas 'cell migration', 'cell growth', 'signaling', and 'metabolic/biosynthetic processes' categories were enriched for GE. For blood vessels, categories such as 'focal adhesion', 'actin cytoskeleton', 'cell junction', 'cell differentiation and development' were found as overrepresented, while for stromal samples, most DEGs were assigned to 'extracellular matrix', 'gap junction', and 'ER to Golgi vesicles'. CONCLUSIONS The localization of differential gene expression to different endometrial cell types provided a significantly improved view on the regulation of biological processes involved in conceptus implantation, such as the control of uterine fluid secretion, trophoblast attachment, growth regulation by Wnt signaling and other signaling pathways, as well as the modulation of the maternal immune system.
Collapse
Affiliation(s)
- Shuqin Zeng
- Genetics and Functional Genomics, Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse Faculty, University of Zurich, Eschikon 27 AgroVet-Strickhof, Zurich, Switzerland
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Lindau, ZH 8315 Switzerland
| | - Susanne E. Ulbrich
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Lindau, ZH 8315 Switzerland
| | - Stefan Bauersachs
- Genetics and Functional Genomics, Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse Faculty, University of Zurich, Eschikon 27 AgroVet-Strickhof, Zurich, Switzerland
| |
Collapse
|
231
|
Joshi S, De Angelis PM, Zucknick M, Schjølberg AR, Andersen SN, Clausen OPF. Role of the Wnt signaling pathway in keratoacanthoma. Cancer Rep (Hoboken) 2019; 3:e1219. [PMID: 32672002 DOI: 10.1002/cnr2.1219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Keratoacanthoma (KA) has a unique life cycle of rapid growth and spontaneous regression that shows similarities to the hair follicle cycle, which involves an active Wnt signaling during physiological regeneration. We analyzed the expression of the Wnt signaling proteins β-catenin, Lef1, Sox9, and Cyclin D1 in young and old human KAs to investigate a possible role for Wnt signaling in KAs. AIM To investigate the role of the Wnt/β-catenin signaling pathway in human KAs. METHODS AND RESULTS Formalin-fixed, paraffin-embedded tissue samples of 67 KAs were analyzed for protein expression using immunohistochemistry. The majority of KAs were positive for Sox9 and Cyclin D1 but not for nuclear-localized β-catenin or Lef-1. No significant differences in protein expressions were seen between young and old KAs. However, we found a significant association between Ki67 and Cyclin D1 proteins (P= .008). CONCLUSIONS The Wnt signaling pathway does not appear to play a significant role in the biogenesis of human KA. Sox9 overexpression may be indicative of inhibition of Wnt signaling. Sox-9 and Cyclin D1 are proliferation markers that are most likely transactivated by alternate signaling pathways.
Collapse
Affiliation(s)
- Sarita Joshi
- Department of Pathology, Rikshospitalet, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Paula M De Angelis
- Department of Pathology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Manuela Zucknick
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Aasa R Schjølberg
- Department of Pathology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Solveig Norheim Andersen
- Institute of Clinical Medicine, Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | | |
Collapse
|
232
|
Wnt-11 Expression Promotes Invasiveness and Correlates with Survival in Human Pancreatic Ductal Adeno Carcinoma. Genes (Basel) 2019; 10:genes10110921. [PMID: 31718047 PMCID: PMC6895970 DOI: 10.3390/genes10110921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/26/2019] [Accepted: 11/05/2019] [Indexed: 01/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest forms of cancer, proving difficult to manage clinically. Wnt-11, a developmentally regulated gene producing a secreted protein, has been associated with various carcinomas but has not previously been studied in PDAC. The present study aimed to elucidate these aspects first in vitro and then in a clinical setting in vivo. Molecular analyses of Wnt-11 expression as well as other biomarkers involved qRT-PCR, RNA-seq and siRNA. Proliferation was measured by MTT; invasiveness was quantified by Boyden chamber (Matrigel) assay. Wnt-11 mRNA was present in three different human PDAC cell lines. Wnt-11 loss affected epithelial-mesenchymal transition and expression of neuronal and stemness biomarkers associated with metastasis. Indeed, silencing Wnt-11 in Panc-1 cells significantly inhibited their Matrigel invasiveness without affecting their proliferative activity. Consistently with the in vitro data, human biopsies of PDAC showed significantly higher Wnt-11 mRNA levels compared with matched adjacent tissues. Expression was significantly upregulated during PDAC progression (TNM stage I to II) and maintained (TNM stages III and IV). Wnt-11 is expressed in PDAC in vitro and in vivo and plays a significant role in the pathophysiology of the disease; this evidence leads to the conclusion that Wnt-11 could serve as a novel, functional biomarker PDAC.
Collapse
|
233
|
Zhuo C, Xun Z, Hou W, Ji F, Lin X, Tian H, Zheng W, Chen M, Liu C, Wang W, Chen C. Surprising Anticancer Activities of Psychiatric Medications: Old Drugs Offer New Hope for Patients With Brain Cancer. Front Pharmacol 2019; 10:1262. [PMID: 31695618 PMCID: PMC6817617 DOI: 10.3389/fphar.2019.01262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022] Open
Abstract
Despite decades of research and major efforts, malignant brain tumors remain among the deadliest of all cancers. Recently, an increasing number of psychiatric drugs has been proven to possess suppressing activities against brain tumors, and rapid progress has been made in understanding the potential mechanisms of action of these drugs. In particular, the traditional mood stabilizer valproic acid, the widely used antidepressants fluoxetine and escitalopram oxalate, and the atypical psychiatric drug aripiprazole have demonstrated promise for application in brain tumor treatment strategies through multiple lines of laboratory, preclinical, and clinical evidence. The unexpected discovery of the anticancer properties of these drugs has ignited interest in the repurposing of other psychiatric drugs to combat brain cancer. In this review, we synthesize recent progress in understanding the potential molecular mechanisms underlying the brain cancer-killing activities of representative psychiatric drugs. We also identify key limitations in the repurposing of these medications that must be overcome to enhance our ability to successfully prevent and treat brain cancer, especially in the most vulnerable groups of patients, such as children and adolescents, pregnant women, and those with unfavorable genetic variants. Moreover, we propose perspectives that may guide future research and provide long-awaited new hope to patients with brain cancer and their families.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China.,Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China.,Department of China-Canada Biological Psychiatry Lab, Xiamen Xianyue Hospital, Xiamen, China.,Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Zhiyuan Xun
- Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Weihong Hou
- Department of Biochemistry and Molecular Biology, Zhengzhou University, Zhengzhou, China.,Department of Biology, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Feng Ji
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China
| | - Xiaodong Lin
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Hongjun Tian
- Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Weifang Zheng
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Min Chen
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China
| | - Chuanxin Liu
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China
| | - Wenqiang Wang
- Department of China-Canada Biological Psychiatry Lab, Xiamen Xianyue Hospital, Xiamen, China
| | - Ce Chen
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| |
Collapse
|
234
|
Dzobo K, Thomford NE, Senthebane DA. Targeting the Versatile Wnt/β-Catenin Pathway in Cancer Biology and Therapeutics: From Concept to Actionable Strategy. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 23:517-538. [PMID: 31613700 DOI: 10.1089/omi.2019.0147] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This expert review offers a critical synthesis of the latest insights and approaches at targeting the Wnt/β-catenin pathway in various cancers such as colorectal cancer, melanoma, leukemia, and breast and lung cancers. Notably, from organogenesis to cancer, the Wnt/β-catenin signaling displays varied and highly versatile biological functions in animals, with virtually all tissues requiring the Wnt/β-catenin signaling in one way or the other. Aberrant expression of the members of the Wnt/β-catenin has been implicated in many pathological conditions, particularly in human cancers. Mutations in the Wnt/β-catenin pathway genes have been noted in diverse cancers. Biochemical and genetic data support the idea that inhibition of Wnt/β-catenin signaling is beneficial in cancer therapeutics. The interaction of this important pathway with other signaling systems is also noteworthy, but remains as an area for further research and discovery. In addition, formation of different complexes by components of the Wnt/β-catenin pathway and the precise roles of these complexes in the cytoplasmic milieu are yet to be fully elucidated. This article highlights the latest medical technologies in imaging, single-cell omics, use of artificial intelligence (e.g., machine learning techniques), genome sequencing, quantum computing, molecular docking, and computational softwares in modeling interactions between molecules and predicting protein-protein and compound-protein interactions pertinent to the biology and therapeutic value of the Wnt/β-catenin signaling pathway. We discuss these emerging technologies in relationship to what is currently needed to move from concept to actionable strategies in translating the Wnt/β-catenin laboratory discoveries to Wnt-targeted cancer therapies and diagnostics in the clinic.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nicholas Ekow Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Dimakatso A Senthebane
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
235
|
Hansen PJ, Tríbulo P. Regulation of present and future development by maternal regulatory signals acting on the embryo during the morula to blastocyst transition - insights from the cow. Biol Reprod 2019; 101:526-537. [PMID: 31220231 PMCID: PMC8127039 DOI: 10.1093/biolre/ioz030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/18/2019] [Indexed: 12/18/2022] Open
Abstract
The preimplantation embryo has a remarkable ability to execute its developmental program using regulatory information inherent within itself. Nonetheless, the uterine environment is rich in cell signaling molecules termed embryokines that act on the embryo during the morula-to-blastocyst transition, promoting blastocyst formation and programming the embryo for subsequent developmental events. Programming can not only affect developmental processes important for continuance of development in utero but also affect characteristics of the offspring during postnatal life. Given the importance of embryokines for regulation of embryonic development, it is likely that some causes of infertility involve aberrant secretion of embryokines by the uterus. Embryokines found to regulate development of the bovine embryo include insulin-like growth factor 1, colony stimulating factor 2 (CSF2), and dickkopf WNT signaling pathway inhibitor 1. Embryo responses to CSF2 exhibit sexual dimorphism, suggesting that sex-specific programming of postnatal function is caused by maternal signals acting on the embryo during the preimplantation period that regulate male embryos differently than female embryos.
Collapse
Affiliation(s)
- Peter J Hansen
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Paula Tríbulo
- Instituto de Reproducción Animal Córdoba (IRAC), Zona Rural General Paz, Córdoba, Argentina
| |
Collapse
|
236
|
Lang CMR, Chan CK, Veltri A, Lien WH. Wnt Signaling Pathways in Keratinocyte Carcinomas. Cancers (Basel) 2019; 11:cancers11091216. [PMID: 31438551 PMCID: PMC6769728 DOI: 10.3390/cancers11091216] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 12/12/2022] Open
Abstract
The skin functions as a barrier between the organism and the surrounding environment. Direct exposure to external stimuli and the accumulation of genetic mutations may lead to abnormal cell growth, irreversible tissue damage and potentially favor skin malignancy. Skin homeostasis is coordinated by an intricate signaling network, and its dysregulation has been implicated in the development of skin cancers. Wnt signaling is one such regulatory pathway orchestrating skin development, homeostasis, and stem cell activation. Aberrant regulation of Wnt signaling cascades not only gives rise to tumor initiation, progression and invasion, but also maintains cancer stem cells which contribute to tumor recurrence. In this review, we summarize recent studies highlighting functional evidence of Wnt-related oncology in keratinocyte carcinomas, as well as discussing preclinical and clinical approaches that target oncogenic Wnt signaling to treat cancers. Our review provides valuable insight into the significance of Wnt signaling for future interventions against keratinocyte carcinomas.
Collapse
Affiliation(s)
| | - Chim Kei Chan
- de Duve Institute, Université catholique de Louvain, Brussels 1200, Belgium
| | - Anthony Veltri
- de Duve Institute, Université catholique de Louvain, Brussels 1200, Belgium
| | - Wen-Hui Lien
- de Duve Institute, Université catholique de Louvain, Brussels 1200, Belgium.
| |
Collapse
|
237
|
5-Azacytidine and trichostatin A enhance the osteogenic differentiation of bone marrow mesenchymal stem cells isolated from steroid-induced avascular necrosis of the femoral head in rabbit. J Biosci 2019. [DOI: 10.1007/s12038-019-9901-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
238
|
Abstract
Developmental signaling pathways control a vast array of biological processes during embryogenesis and in adult life. The WNT pathway was discovered simultaneously in cancer and development. Recent advances have expanded the role of WNT to a wide range of pathologies in humans. Here, we discuss the WNT pathway and its role in human disease and some of the advances in WNT-related treatments.
Collapse
|
239
|
Karvonen H, Barker H, Kaleva L, Niininen W, Ungureanu D. Molecular Mechanisms Associated with ROR1-Mediated Drug Resistance: Crosstalk with Hippo-YAP/TAZ and BMI-1 Pathways. Cells 2019; 8:cells8080812. [PMID: 31382410 PMCID: PMC6721603 DOI: 10.3390/cells8080812] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
Signaling via the Wnt-related receptor tyrosine kinase-like orphan receptor 1 (ROR1) triggers tumorigenic features associated with cancer stem cells (CSCs) and epithelial-mesenchymal transition (EMT), while aberrant expression of ROR1 is strongly linked to advanced disease progression and chemoresistance. Several recent studies have shown that Wnt5a binding to ROR1 promotes oncogenic signaling by activating multiple pathways such as RhoA/Rac1 GTPases and PI3K/AKT, which in turn could induce transcriptional coactivator YAP/TAZ or polycomb complex protein BMI-1 signaling, respectively, to sustain stemness, metastasis and ultimately drug-resistance. These data point towards a new feedback loop during cancer development, linking Wnt5a-ROR1 signaling activation to YAP/TAZ or BMI-1 upregulation that could play an important role in disease progression and treatment resistance. This review focuses on the crosstalk between Wnt5a-ROR1 and YAP/TAZ or the BMI-1 signaling network, together with the current advancements in targeted strategies for ROR1-positive cancers.
Collapse
Affiliation(s)
- Hanna Karvonen
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland
| | - Harlan Barker
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Laura Kaleva
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland
| | - Wilhelmiina Niininen
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland
| | - Daniela Ungureanu
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland.
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland.
| |
Collapse
|
240
|
Iyer LM, Nagarajan S, Woelfer M, Schoger E, Khadjeh S, Zafiriou MP, Kari V, Herting J, Pang ST, Weber T, Rathjens FS, Fischer TH, Toischer K, Hasenfuss G, Noack C, Johnsen SA, Zelarayán LC. A context-specific cardiac β-catenin and GATA4 interaction influences TCF7L2 occupancy and remodels chromatin driving disease progression in the adult heart. Nucleic Acids Res 2019; 46:2850-2867. [PMID: 29394407 PMCID: PMC5887416 DOI: 10.1093/nar/gky049] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/18/2018] [Indexed: 12/17/2022] Open
Abstract
Chromatin remodelling precedes transcriptional and structural changes in heart failure. A body of work suggests roles for the developmental Wnt signalling pathway in cardiac remodelling. Hitherto, there is no evidence supporting a direct role of Wnt nuclear components in regulating chromatin landscapes in this process. We show that transcriptionally active, nuclear, phosphorylated(p)Ser675-β-catenin and TCF7L2 are upregulated in diseased murine and human cardiac ventricles. We report that inducible cardiomyocytes (CM)-specific pSer675-β-catenin accumulation mimics the disease situation by triggering TCF7L2 expression. This enhances active chromatin, characterized by increased H3K27ac and TCF7L2 occupancies to cardiac developmental and remodelling genes in vivo. Accordingly, transcriptomic analysis of β-catenin stabilized hearts shows a strong recapitulation of cardiac developmental processes like cell cycling and cytoskeletal remodelling. Mechanistically, TCF7L2 co-occupies distal genomic regions with cardiac transcription factors NKX2–5 and GATA4 in stabilized-β-catenin hearts. Validation assays revealed a previously unrecognized function of GATA4 as a cardiac repressor of the TCF7L2/β-catenin complex in vivo, thereby defining a transcriptional switch controlling disease progression. Conversely, preventing β-catenin activation post-pressure-overload results in a downregulation of these novel TCF7L2-targets and rescues cardiac function. Thus, we present a novel role for TCF7L2/β-catenin in CMs-specific chromatin modulation, which could be exploited for manipulating the ubiquitous Wnt pathway.
Collapse
Affiliation(s)
- Lavanya M Iyer
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Sankari Nagarajan
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,Cancer Research UK (CRUK-CI), Cambridge CB2 0RE, UK
| | - Monique Woelfer
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Eric Schoger
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Sara Khadjeh
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Maria Patapia Zafiriou
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Vijayalakshmi Kari
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Jonas Herting
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Sze Ting Pang
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Tobias Weber
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Franziska S Rathjens
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Thomas H Fischer
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Karl Toischer
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Gerd Hasenfuss
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Claudia Noack
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Steven A Johnsen
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Laura C Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| |
Collapse
|
241
|
Li J, Xue K, Zheng Y, Wang Y, Xu C. RORA Overexpression Alleviates Nasal Mucosal Injury and Enhances Red Blood Cell Immune Adhesion Function in a Mouse Model of Allergic Rhinitis via Inactivation of the Wnt/β-Catenin Signaling Pathway. Int Arch Allergy Immunol 2019; 180:79-90. [PMID: 31340215 DOI: 10.1159/000500637] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/26/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In this study, we examined whether RORA (retinoic acid receptor-related orphan receptor alpha) was capable of alleviating the progression of allergic rhinitis (AR). METHODS In order to elucidate the possible effects of RORA and the regulatory mechanism between RORA and the Wnt/β-catenin signaling pathway, mouse AR models were established and treated with RORA vector, siRNA against RORA, or the Wnt/β-catenin pathway inhibitor WIF-1. Subsequently, the serum levels of inflammatory cytokines (IgE, INF-γ, IL-1β, IL-4, and IL-17), red blood cell (RBC) immune adhesion function, the levels of RORA, β-catenin, and GSK3β, as well as the extent of β-catenin and GSK-3β phosphorylation were evaluated and measured. RESULTS The OVA-induced AR mouse model exhibited obvious nasal mucosal injury and inflammatory cell infiltration. RORA overexpression or the inactivation of the Wnt/β-catenin signaling pathway was uncovered as a way to ameliorate nasal mucosal injury and eosinophil infiltration of the OVA-induced AR mouse model. On the other hand, it reduced the number of eosinophils and mast cells, which also resulted in downregulated expression of IgE, INF-γ, IL-1β, IL-4, IL-17, β-catenin, and GSK-3β. Moreover, this led to a decreased extent of β-catenin and GSK-3β phosphorylation, while the rates of C3b receptor rosette and Ic rosette were elevated. CONCLUSION Taken together, the key findings provided evidence suggesting that the elevated RORA could potentially alleviate nasal mucosal injury and simultaneously enhance RBC immune adhesion function through the inhibition of the Wnt/β-catenin signaling pathway activation in an OVA-induced AR mouse model. This emphasizes a novel therapeutic target for the treatment of AR.
Collapse
Affiliation(s)
- Jinqiu Li
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Kai Xue
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yan Zheng
- Department of Anesthesiology, China-Japan Union Hospital, Changchun, China
| | - Yinan Wang
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, China
| | - Chengbi Xu
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China,
| |
Collapse
|
242
|
Postlethwait JH, Navajas Acedo J, Piotrowski T. Evolutionary Origin and Nomenclature of Vertebrate Wnt11-Family Genes. Zebrafish 2019; 16:469-476. [PMID: 31295059 DOI: 10.1089/zeb.2019.1760] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
To adequately connect zebrafish medical models to human biology, it is essential that gene nomenclature reflects gene orthology. Analysis of gene phylogenies and conserved syntenies shows that the zebrafish gene currently called wnt11 (ENSDARG00000004256, ZFIN ID: ZDB-GENE-990603-12) is not the ortholog of the human gene called WNT11 (ENSG00000085741); instead, the gene currently called wnt11r (ENSDARG00000014796, ZFIN ID: ZDB-GENE-980526-249) is the zebrafish ortholog of human WNT11. Genomic analysis of Wnt11-family genes suggests a model for the birth of Wnt11-family gene ohnologs in genome duplication events, provides a mechanism for the death of a Wnt11-family ohnolog in mammals after they diverged from birds, and suggests revised nomenclature to better connect teleost disease models to human biology.
Collapse
|
243
|
Liu D, Li S, Cui Y, Tong H, Li S, Yan Y. Podocan affects C2C12 myogenic differentiation by enhancing Wnt/β-catenin signaling. J Cell Physiol 2019; 234:11130-11139. [PMID: 30652305 DOI: 10.1002/jcp.27763] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022]
Abstract
Podocan, a small leucine-rich repeat protein, is a negative regulator of cell proliferation. In this study, we demonstrated that podocan is involved in the differentiation of C2C12 murine myoblasts. Podocan expression increased with the progression of C2C12 differentiation. As expect, siRNA-mediated podocan knockdown inhibited C2C12 differentiation, as indicated by inhibition of MYOG, MYH2, and desmin expression, as well as reductions in the differentiation and fusion indices. Overexpression of podocan using dCas9 technology promoted C2C12 cell differentiation. In addition, supplementation of culture medium with podocan influenced C2C12 differentiation. Podocan knockdown reduced Wnt/β-catenin signaling, characterized by a reduction in the nuclear translocation of β-catenin, whereas podocan overexpression had the opposite effect. Furthermore, treatment with XAV939, an inhibitor of Wnt/β-catenin, reduced the podocan-mediated promotion of C2C12 differentiation. Induction of muscle injury in mice by bupivacaine administration suggested that podocan may play a role in muscle regeneration. In summary, our results suggest that podocan is required for normal C2C12 differentiation and that its role in myogenesis is mediated by the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Dan Liu
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shuang Li
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yafeng Cui
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Huili Tong
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shufeng Li
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yunqin Yan
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| |
Collapse
|
244
|
Bai L, Gao S, Sun H, Zhao X, Yang L, Hu H, Sun J, Jiang W. Effects of Wnt10b on dermal papilla cells via the canonical Wnt/β-catenin signalling pathway in the Angora rabbit. J Anim Physiol Anim Nutr (Berl) 2019; 103:1602-1609. [PMID: 31241227 DOI: 10.1111/jpn.13141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 01/19/2023]
Abstract
Wnt10b is a member of Wnt family that plays a variety of roles in biological functions, including those in the development of hair follicles. To investigate the effect of Wnt10b on hair growth in the Angora rabbit and to determine the underlying molecular mechanism, we cultured dermal papilla (DP) cells with exogenous Wnt10b in vitro. We observed the expressions of downstream critical gene β-catenin and lymphoid enhancer-binding factor 1 (LEF1) in Wnt/β-catenin pathway. The levels of β-catenin mRNA and protein were higher in the Wnt10b group of DP cells than in the Control group, and the mRNA level of LEF1 in the Wnt10b group was higher than in the Control group. Moreover, translocation of β-catenin from cytoplasm to nucleus was activated in the Wnt10b group. Furthermore, the mRNA levels of the hair follicle-regulatory genes, insulin-like growth factor-1 (IGF-1) and alkaline phosphatase (ALP), and the protein activity of ALP was also upregulated in the Wnt10b group compared to their corresponding levels in the Control group. These data suggest that Wnt10b could activate the canonical Wnt/β-catenin signalling pathway to induce DP cells in the Angora rabbit. In addition, the proliferation of DP cells was significantly promoted when cultured with Wnt10b for 48 and 72 hr, suggesting that Wnt10b plays a pivotal role in the proliferation and maintenance of DP cells in vitro. In conclusion, this study demonstrates that Wnt10b may promote hair follicle growth in Angora rabbit through the canonical Wnt/β-catenin signalling pathway that promotes the proliferation of DP cells.
Collapse
Affiliation(s)
- Liya Bai
- Shandong Academy of Agricultural Sciences, Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Jinan, China
| | - Shuxia Gao
- Shandong Academy of Agricultural Sciences, Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Jinan, China
| | - Haitao Sun
- Shandong Academy of Agricultural Sciences, Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Jinan, China
| | - Xueyan Zhao
- Shandong Academy of Agricultural Sciences, Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Jinan, China
| | - Liping Yang
- Shandong Academy of Agricultural Sciences, Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Jinan, China
| | - Hongmei Hu
- Shandong Academy of Agricultural Sciences, Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Jinan, China
| | - Jie Sun
- Xingtai Bureau of Agriculture and Rural Affairs, Xingtai, China
| | - Wenxue Jiang
- Shandong Academy of Agricultural Sciences, Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Jinan, China
| |
Collapse
|
245
|
Lv MY, Shi CJ, Pan FF, Shao J, Feng L, Chen G, Ou C, Zhang JF, Fu WM. Urolithin B suppresses tumor growth in hepatocellular carcinoma through inducing the inactivation of Wnt/β-catenin signaling. J Cell Biochem 2019; 120:17273-17282. [PMID: 31218741 DOI: 10.1002/jcb.28989] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/16/2022]
Abstract
Consumption of dietary ellagitannins (ETs) has been proven to benefit multiple chronic health disorders including cancers and cardiovascular diseases. Urolithins, gut microbiota metabolites derived from ETs, are considered as the molecules responsible for these health effects. Previous studies have demonstrated that urolithins exhibit antiproliferative effects on prostate, breast, and colon cancers. However, as for hepatocellular carcinoma (HCC), it remains elusive. Herein, we aim to investigate the function of urolithin B (UB), a member of urolithins family, in HCC. The effects of UB on cell viability, cell cycle and apoptosis were evaluated in HCC cells, and we found UB could inhibit the proliferation of HCC cells, which resulted from cell cycle arrest and apoptosis. Furthermore, UB could increase phosphorylated β-catenin expression and block its translocation from nuclear to cytoplasm, thus inducing the inactivation of Wnt/β-catenin signaling. Using a xenograft mice model, UB was found to suppress tumor growth in vivo. In conclusion, our data demonstrated that UB could inhibit the proliferation of HCC cells in vitro and in vivo via inactivating Wnt/β-catenin signaling, suggesting UB could be a promising candidate in the development of anticancer drugs targeting HCC.
Collapse
Affiliation(s)
- Min-Yi Lv
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Chuan-Jian Shi
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Fei-Fei Pan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Jiang Shao
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Lu Feng
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Guoqin Chen
- Department of Central Hospital of Panyu, Cardiovascular Medicine, Guangzhou, People's Republic of China
| | - Caiwen Ou
- Zhujiang Hospital, Southern Medical University, Key Laboratory of Construction and Detection of Guangdong Province, Guangdong Province Center of Biomedical Engineering for Cardiovascular Diseases, No. 1023, Shatai Nan Road, Guangzhou, People's Republic of China
| | - Jin-Fang Zhang
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Wei-Ming Fu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
246
|
Liao YJ, Yin XL, Deng Y, Peng XW. PRC1 gene silencing inhibits proliferation, invasion, and angiogenesis of retinoblastoma cells through the inhibition of the Wnt/β-catenin signaling pathway. J Cell Biochem 2019; 120:16840-16852. [PMID: 31144388 DOI: 10.1002/jcb.28942] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 04/11/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022]
Abstract
Retinoblastoma is an ocular malignancy occurring in childhood. The current study evaluates the ability of silenced PRC1 on retinoblastoma cell proliferation, and angiogenesis via the Wnt/β-catenin signaling pathway. A total of 36 cases of retinoblastoma tissues (n = 36) and normal retinal tissues (n = 10) were selected in the current study. Retinoblastoma cells presenting with the high PRC1 messenger RNA (mRNA) expression were selected among the WERI-Rb-1, HXO-RB44, Y79, SO-Rb50, and SO-Rb70 cells lines, and were transfected with siRNA-PRC1 and LiCl (the activator of the Wnt/β-catenin pathway). The expressions of PRC1, VEGF, Wnt1, β-catenin, CyclinD1, extent of β-catenin, and GSK-3β phosphorylation were evaluated. Cell proliferation, cell-cycle distribution, and cell invasion of retinoblastoma cells were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, flow cytometry, and Transwell assay. The angiogenesis of retinoblastoma cells was detected by tube formation assay. HXO-RB44 and WERI-Rb-1 cells were selected owing to the highest PRC1 mRNA expression. Meanwhile, PRC2 gene silencing presented lower expression levels of PRC1, VEGF, Wnt1, β-catenin, CyclinD1, extent of β-catenin and GSK-3β phosphorylation, decreased proliferation and invasion abilities, extended G0/G1 phase, and shortened S and G2/M phases of HXO-RB44 and WERI-Rb-1 cells, suggesting the silenced PRC2 inactivated Wnt/β-catenin pathway, so as to further restrain the retinoblastoma cell proliferation, invasion, and angiogenesis. These results support the view that PRC1 gene silencing could suppress the proliferation, and angiogenesis of retinoblastoma cells by repressing the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yu-Jun Liao
- Department of Pediatric Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Xiao-Long Yin
- Department of Pediatric Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Yan Deng
- Department of Pediatric Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Xiao-Wei Peng
- Department of Pediatric Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| |
Collapse
|
247
|
Guttuso T, Andrzejewski KL, Lichter DG, Andersen JK. Targeting kinases in Parkinson's disease: A mechanism shared by LRRK2, neurotrophins, exenatide, urate, nilotinib and lithium. J Neurol Sci 2019; 402:121-130. [PMID: 31129265 DOI: 10.1016/j.jns.2019.05.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/16/2022]
Abstract
Several kinases have been implicated in the pathogenesis of Parkinson's disease (PD), most notably leucine-rich repeat kinase 2 (LRRK2), as LRRK2 mutations are the most common genetic cause of a late-onset parkinsonism that is clinically indistinguishable from sporadic PD. More recently, several other kinases have emerged as promising disease-modifying targets in PD based on both preclinical studies and clinical reports on exenatide, the urate precursor inosine, nilotinib and lithium use in PD patients. These kinases include protein kinase B (Akt), glycogen synthase kinases-3β and -3α (GSK-3β and GSK-3α), c-Abelson kinase (c-Abl) and cyclin-dependent kinase 5 (cdk5). Activities of each of these kinases are involved either directly or indirectly in phosphorylating tau or increasing α-synuclein levels, intracellular proteins whose toxic oligomeric forms are strongly implicated in the pathogenesis of PD. GSK-3β, GSK-3α and cdk5 are the principle kinases involved in phosphorylating tau at sites critical for the formation of tau oligomers. Exenatide analogues, urate, nilotinib and lithium have been shown to affect one or more of the above kinases, actions that can decrease the formation and increase the clearance of intraneuronal phosphorylated tau and α-synuclein. Here we review the current preclinical and clinical evidence supporting kinase-targeting agents as potential disease-modifying therapies for PD patients enriched with these therapeutic targets and incorporate LRRK2 physiology into this novel model.
Collapse
Affiliation(s)
- Thomas Guttuso
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America.
| | - Kelly L Andrzejewski
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America.
| | - David G Lichter
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America.
| | - Julie K Andersen
- The Buck Institute for Research on Aging, Novato, CA, United States of America.
| |
Collapse
|
248
|
Park SM, Jang HJ, Lee JH. Roles of Primary Cilia in the Developing Brain. Front Cell Neurosci 2019; 13:218. [PMID: 31139054 PMCID: PMC6527876 DOI: 10.3389/fncel.2019.00218] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/30/2019] [Indexed: 01/07/2023] Open
Abstract
Essential to development, primary cilia are microtubule-based cellular organelles that protrude from the surface of cells. Acting as cellular antenna, primary cilia play central roles in transducing or regulating several signaling pathways, including Sonic hedgehog (Shh) and Wnt signaling. Defects in primary cilia contribute to a group of syndromic disorders known as “ciliopathies” and can adversely affect development of the brain and other essential organs, including the kidneys, eyes, and liver. The molecular mechanisms of how defective primary cilia contribute to neurological defects, however, remain poorly understood. In this mini review, we summarize recent advances in understanding of the interactions between primary cilia and signaling pathways essential to cellular homeostasis and brain development.
Collapse
Affiliation(s)
- Sang Min Park
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Hee Jin Jang
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| |
Collapse
|
249
|
Zhang T, Chen H, Zhou Y, Dong W, Cai H, Tan WS. Cooperation of FGF/MEK/ERK and Wnt/β-catenin pathway regulators to promote the proliferation and pluripotency of mouse embryonic stem cells in serum- and feeder-free conditions. BIORESOUR BIOPROCESS 2019. [DOI: 10.1186/s40643-019-0249-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
250
|
Kerekes K, Bányai L, Trexler M, Patthy L. Structure, function and disease relevance of Wnt inhibitory factor 1, a secreted protein controlling the Wnt and hedgehog pathways. Growth Factors 2019; 37:29-52. [PMID: 31210071 DOI: 10.1080/08977194.2019.1626380] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Wnts and Hedgehogs (Hh) are large, lipid-modified extracellular morphogens that play key roles in embryonic development and stem cell proliferation of Metazoa. Both morphogens signal through heptahelical Frizzled-type receptors of the G-Protein Coupled Receptor family and there are several other similarities that suggest a common evolutionary origin of the Hh and Wnt pathways. There is evidence that the secreted protein, Wnt inhibitory factor 1 (WIF1) modulates the activity of both Wnts and Hhs and may thus contribute to the intertwining of these pathways. In this article, we review the structure, evolution, molecular interactions and functions of WIF1 with major emphasis on its role in carcinogenesis.
Collapse
Affiliation(s)
- Krisztina Kerekes
- a Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - László Bányai
- a Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - Mária Trexler
- a Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - László Patthy
- a Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| |
Collapse
|