151
|
Macarak E, Rosenbloom J. The Pathogenesis of Intraabdominal Adhesions: Similarities and Differences to Luminal Fibrosis. FIBROSTENOTIC INFLAMMATORY BOWEL DISEASE 2018:319-346. [DOI: 10.1007/978-3-319-90578-5_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
152
|
Amini-Nik S, Yousuf Y, Jeschke MG. Scar management in burn injuries using drug delivery and molecular signaling: Current treatments and future directions. Adv Drug Deliv Rev 2018; 123:135-154. [PMID: 28757325 PMCID: PMC5742037 DOI: 10.1016/j.addr.2017.07.017] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
In recent decades, there have been tremendous improvements in burn care that have allowed patients to survive severe burn injuries that were once fatal. However, a major limitation of burn care currently is the development of hypertrophic scars in approximately 70% of patients. This significantly decreases the quality of life for patients due to the physical and psychosocial symptoms associated with scarring. Current approaches to manage scarring include surgical techniques and non-surgical methods such as laser therapy, steroid injections, and compression therapy. These treatments are limited in their effectiveness and regularly fail to manage symptoms. As a result, the development of novel treatments that aim to improve outcomes and quality of life is imperative. Drug delivery that targets the molecular cascades of wound healing to attenuate or prevent hypertrophic scarring is a promising approach that has therapeutic potential. In this review, we discuss current treatments for scar management after burn injury, and how drug delivery targeting molecular signaling can lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Saeid Amini-Nik
- Sunnybrook Research Institute, Toronto, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada.
| | - Yusef Yousuf
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada
| | - Marc G Jeschke
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada; Department of Immunology, University of Toronto, Toronto, Canada; Ross-Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada.
| |
Collapse
|
153
|
Oliva CA, Montecinos-Oliva C, Inestrosa NC. Wnt Signaling in the Central Nervous System: New Insights in Health and Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 153:81-130. [PMID: 29389523 DOI: 10.1016/bs.pmbts.2017.11.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Since its discovery, Wnt signaling has been shown to be one of the most crucial morphogens in development and during the maturation of central nervous system. Its action is relevant during the establishment and maintenance of synaptic structure and neuronal function. In this chapter, we will discuss the most recent evidence on these aspects, and we will explore the evidence that involves Wnt signaling on other less known functions, such as in adult neurogenesis, in the generation of oscillatory neural rhythms, and in adult behavior. The dysfunction of Wnt signaling at different levels will be also discussed, in particular in those aspects that have been found to be linked with several neurodegenerative diseases and neurological disorders. Finally, we will address the possibility of Wnt signaling manipulation to treat those pathophysiological aspects.
Collapse
Affiliation(s)
- Carolina A Oliva
- Center for Aging and Regeneration (CARE-UC), Pontifical Catholic University of Chile, Santiago, Chile
| | - Carla Montecinos-Oliva
- Center for Aging and Regeneration (CARE-UC), Pontifical Catholic University of Chile, Santiago, Chile; Interdisciplinary Institute for Neuroscience (IINS), University of Bordeaux, Bordeaux, France
| | - Nibaldo C Inestrosa
- Center for Aging and Regeneration (CARE-UC), Pontifical Catholic University of Chile, Santiago, Chile; Center for Healthy Brain Ageing, University of New South Wales, Sydney, NSW, Australia; Center of Excellence in Biomedicine of Magallanes (CEBIMA), University of Magallanes, Punta Arenas, Chile.
| |
Collapse
|
154
|
|
155
|
Kagey MH, He X. Rationale for targeting the Wnt signalling modulator Dickkopf-1 for oncology. Br J Pharmacol 2017; 174:4637-4650. [PMID: 28574171 PMCID: PMC5727329 DOI: 10.1111/bph.13894] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/12/2017] [Accepted: 05/19/2017] [Indexed: 12/15/2022] Open
Abstract
Wnt signalling is a fundamental pathway involved in embryonic development and adult tissue homeostasis. Mutations in the pathway frequently lead to developmental defects and cancer. As such, therapeutic intervention of this pathway has generated tremendous interest. Dickkopf-1 (DKK1) is a secreted inhibitor of β-catenin-dependent Wnt signalling and was originally characterized as a tumour suppressor based on the prevailing view that Wnt signalling promotes cancer pathogenesis. However, DKK1 appears to increase tumour growth and metastasis in preclinical models and its elevated expression correlates with a poor prognosis in a range of cancers, indicating that DKK1 has more complex cellular and biological functions than originally appreciated. Here, we review current evidence for the cancer-promoting activity of DKK1 and recent insights into the effects of DKK1 on signalling pathways in both cancer and immune cells. We discuss the rationale and promise of targeting DKK1 for oncology. LINKED ARTICLES This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc.
Collapse
Affiliation(s)
| | - Xi He
- The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Department of NeurologyHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
156
|
Driehuis E, Clevers H. WNT signalling events near the cell membrane and their pharmacological targeting for the treatment of cancer. Br J Pharmacol 2017; 174:4547-4563. [PMID: 28244067 PMCID: PMC5727251 DOI: 10.1111/bph.13758] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/16/2017] [Accepted: 02/18/2017] [Indexed: 12/16/2022] Open
Abstract
WNT signalling is an essential signalling pathway for all multicellular animals. Although first described more than 30 years ago, new components and regulators of the pathway are still being discovered. Considering its importance in both embryonic development and adult homeostasis, it is not surprising that this pathway is often deregulated in human diseases such as cancer. Recently, it became clear that in addition to cytoplasmic components such as β-catenin, other, membrane-bound or extracellular, components of the WNT pathway are also altered in cancer. This review gives an overview of the recent discoveries on WNT signalling events near the cell membrane. Furthermore, membrane-associated components of the WNT pathway, which are more accessible for therapeutic intervention, as well therapeutic approaches that already target those components will be discussed. In this way, we hope to stimulate the development of effective anti-cancer therapies that target this fascinating pathway. LINKED ARTICLES This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc.
Collapse
Affiliation(s)
- Else Driehuis
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW)UtrechtThe Netherlands
- University medical center (UMC)UtrechtThe Netherlands
| | - Hans Clevers
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW)UtrechtThe Netherlands
- University medical center (UMC)UtrechtThe Netherlands
- Princess Maxime Center (PMC)UtrechtThe Netherlands
| |
Collapse
|
157
|
Zhong L, Schivo S, Huang X, Leijten J, Karperien M, Post JN. Nitric Oxide Mediates Crosstalk between Interleukin 1β and WNT Signaling in Primary Human Chondrocytes by Reducing DKK1 and FRZB Expression. Int J Mol Sci 2017; 18:ijms18112491. [PMID: 29165387 PMCID: PMC5713457 DOI: 10.3390/ijms18112491] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/20/2022] Open
Abstract
Interleukin 1 beta (IL1β) and Wingless-Type MMTV Integration Site Family (WNT) signaling are major players in Osteoarthritis (OA) pathogenesis. Despite having a large functional overlap in OA onset and development, the mechanism of IL1β and WNT crosstalk has remained largely unknown. In this study, we have used a combination of computational modeling and molecular biology to reveal direct or indirect crosstalk between these pathways. Specifically, we revealed a mechanism by which IL1β upregulates WNT signaling via downregulating WNT antagonists, DKK1 and FRZB. In human chondrocytes, IL1β decreased the expression of Dickkopf-1 (DKK1) and Frizzled related protein (FRZB) through upregulation of nitric oxide synthase (iNOS), thereby activating the transcription of WNT target genes. This effect could be reversed by iNOS inhibitor 1400W, which restored DKK1 and FRZB expression and their inhibitory effect on WNT signaling. In addition, 1400W also inhibited both the matrix metalloproteinase (MMP) expression and cytokine-induced apoptosis. We concluded that iNOS/NO play a pivotal role in the inflammatory response of human OA through indirect upregulation of WNT signaling. Blocking NO production may inhibit the loss of the articular phenotype in OA by preventing downregulation of the expression of DKK1 and FRZB.
Collapse
Affiliation(s)
- Leilei Zhong
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, The Netherlands.
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Stefano Schivo
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, The Netherlands.
- Formal Methods and Tools, CTIT, University of Twente, 7522 NB Enschede, The Netherlands.
| | - Xiaobin Huang
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, The Netherlands.
| | - Jeroen Leijten
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, The Netherlands.
| | - Marcel Karperien
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, The Netherlands.
| | - Janine N Post
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, The Netherlands.
| |
Collapse
|
158
|
Zhou M, Wu J, Yu Y, Yang Y, Li J, Cui L, Yao W, Liu Y. Polygonum multiflorm alleviates glucocorticoid‑induced osteoporosis and Wnt signaling pathway. Mol Med Rep 2017; 17:970-978. [PMID: 29115514 PMCID: PMC5780178 DOI: 10.3892/mmr.2017.7997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 10/10/2017] [Indexed: 12/18/2022] Open
Abstract
It is known that long-term excessive administration of glucocorticoid (GC) results in osteoporosis. The present study aimed to evaluate the protective effects of Polygonum multiflorm (PM) on the bone tissue of rats with GC-induced osteoporosis (GIO). A total of 90 6-month-old female Sprague Dawley rats (weight range, 190–210 g) were randomly divided into nine groups: Control (normal saline); prednisone (GC; 6 mg·kg−1·d−1; Model); GC plus PMR30 (the 30% ethanol eluent fraction of PM) (H) (400 mg·kg−1·d−1); GC plus PMR30 (M) (200 mg·kg−1·d−1); GC plus PMR30 (L) (100 mg·kg−1·d−1); GC plus PMRF (fat-soluble fraction of PM) (H) (400 mg·kg−1·d−1); GC plus PMRF (M) (200 mg·kg−1·d−1); GC plus PMRF (L) (100 mg·kg−1·d−1); GC plus calcitriol (CAL; 0.045 µg·kg−1·d−1; positive). Rats were administered intragastrically with prednisone and/or the aforementioned extracts for 120 days, and weighed once/week. The serum was collected for detection of biochemical markers. The left tibia was used for bone histomorphometry analysis. The right tibia was prepared for hematoxylin and eosin staining. The left femur was used to analyze the protein expression of dickkopf-1 (DKK1), WNT inhibitory factor 1 (WIF1) and secreted frizzled related protein 4 using western blotting. Long-term excessive treatment of prednisone inhibited the bone formation rate accompanied with a decrease in bone mass, growth plate, body weight, and the level of bone-specific alkaline phosphatase and hydroxyl-terminal propeptide of type I procollagen in the serum. Furthermore, a simultaneously increase in the level of tartrate resistant acid phosphatase-5b and cross-linked carboxy-terminal telopeptide of type I collagen in the serum, in addition to DKK1, and WIF1 protein expression, was observed. PMR30 (M and L) and PMRF (H) groups were able to reduce the negative effects of GC on the bones. PMR30 (M and L) and PMRF (H) dose demonstrated a protective effect of PM on bone tissue in GIO rats. The mechanism underlying the preventive effect of PM for the treatment of GIO may be associated with direct upregulation of the canonical Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Manru Zhou
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Jingkai Wu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Yongjie Yu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Yajun Yang
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Jin Li
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Weimin Yao
- Department of Respiratory Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Yuyu Liu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| |
Collapse
|
159
|
Miller-Kleinhenz J, Guo X, Qian W, Zhou H, Bozeman EN, Zhu L, Ji X, Wang YA, Styblo T, O'Regan R, Mao H, Yang L. Dual-targeting Wnt and uPA receptors using peptide conjugated ultra-small nanoparticle drug carriers inhibited cancer stem-cell phenotype in chemo-resistant breast cancer. Biomaterials 2017; 152:47-62. [PMID: 29107218 DOI: 10.1016/j.biomaterials.2017.10.035] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/15/2017] [Accepted: 10/18/2017] [Indexed: 12/27/2022]
Abstract
Heterogeneous tumor cells, high incidence of tumor recurrence, and decrease in overall survival are the major challenges for the treatment of chemo-resistant breast cancer. Results of our study showed differential chemotherapeutic responses among breast cancer patient derived xenograft (PDX) tumors established from the same patients. All doxorubicin (Dox)-resistant tumors expressed higher levels of cancer stem-like cell biomarkers, including CD44, Wnt and its receptor LRP5/6, relative to Dox-sensitive tumors. To effectively treat resistant tumors, we developed an ultra-small magnetic iron oxide nanoparticle (IONP) drug carrier conjugated with peptides that are dually targeted to Wnt/LRP5/6 and urokinase plasminogen activator receptor (uPAR). Our results showed that simultaneous binding to LRP5/6 and uPAR by the dual receptor targeted IONPs was required to inhibit breast cancer cell invasion. Molecular analysis revealed that the dual receptor targeted IONPs significantly inhibited Wnt/β-catenin signaling and cancer stem-like phenotype of tumor cells, with marked reduction of Wnt ligand, CD44 and uPAR. Systemic administration of the dual targeted IONPs led to nanoparticle-drug delivery into PDX tumors, resulting in stronger tumor growth inhibition compared to non-targeted or single-targeted IONP-Dox in a human breast cancer PDX model. Therefore, co-targeting Wnt/LRP and uPAR using IONP drug carriers is a promising therapeutic approach for effective drug delivery to chemo-resistant breast cancer.
Collapse
Affiliation(s)
- Jasmine Miller-Kleinhenz
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiangxue Guo
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Weiping Qian
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Hongyu Zhou
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Erica N Bozeman
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Lei Zhu
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Xin Ji
- Ocean Nanotech, San Diego, CA, USA
| | | | - Toncred Styblo
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Ruth O'Regan
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Lily Yang
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
160
|
Amyloid β synaptotoxicity is Wnt-PCP dependent and blocked by fasudil. Alzheimers Dement 2017; 14:306-317. [PMID: 29055813 PMCID: PMC5869054 DOI: 10.1016/j.jalz.2017.09.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/13/2017] [Accepted: 09/07/2017] [Indexed: 01/18/2023]
Abstract
Introduction Synapse loss is the structural correlate of the cognitive decline indicative of dementia. In the brains of Alzheimer's disease sufferers, amyloid β (Aβ) peptides aggregate to form senile plaques but as soluble peptides are toxic to synapses. We previously demonstrated that Aβ induces Dickkopf-1 (Dkk1), which in turn activates the Wnt–planar cell polarity (Wnt-PCP) pathway to drive tau pathology and neuronal death. Methods We compared the effects of Aβ and of Dkk1 on synapse morphology and memory impairment while inhibiting or silencing key elements of the Wnt-PCP pathway. Results We demonstrate that Aβ synaptotoxicity is also Dkk1 and Wnt-PCP dependent, mediated by the arm of Wnt-PCP regulating actin cytoskeletal dynamics via Daam1, RhoA and ROCK, and can be blocked by the drug fasudil. Discussion Our data add to the importance of aberrant Wnt signaling in Alzheimer's disease neuropathology and indicate that fasudil could be repurposed as a treatment for the disease. Aβ synaptotoxicity is Dickkopf-1 and Wnt-PCP dependent. The Wnt-PCP pathway drives Aβ-driven synapse loss via RhoA and ROCK. ROCK inhibitor fasudil blocks Aβ-driven synapse loss and cognitive impairment. Fasudil should be assessed for repurposing for Alzheimer's disease.
Collapse
|
161
|
Ma C, Lu YF. Role of Dickkopf-1 in gastrointestinal tumors. Shijie Huaren Xiaohua Zazhi 2017; 25:2615-2620. [DOI: 10.11569/wcjd.v25.i29.2615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
As an endogenous inhibitor of the Wnt signaling pathway, Dickkopf-1 (DKK1) plays an important role in the occurrence and development of tumors. However, the role of DKK1 in different tumor types is different, as it is lowly expressed in some tumor types, but over-expressed in others. In recent years, there have been many studies on the role of DKK1 in gastrointestinal tumors. In this paper, we review the latest progress in the understanding of the role of DKK1 in gastrointestinal tumors.
Collapse
Affiliation(s)
- Chao Ma
- Department of Gastroenterology, the Affiliated Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - Yong-Fu Lu
- Department of Gastroenterology, the Affiliated Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| |
Collapse
|
162
|
Lu Y, Zhao X, Liu Q, Li C, Graves-Deal R, Cao Z, Singh B, Franklin JL, Wang J, Hu H, Wei T, Yang M, Yeatman TJ, Lee E, Saito-Diaz K, Hinger S, Patton JG, Chung CH, Emmrich S, Klusmann JH, Fan D, Coffey RJ. lncRNA MIR100HG-derived miR-100 and miR-125b mediate cetuximab resistance via Wnt/β-catenin signaling. Nat Med 2017; 23:1331-1341. [PMID: 29035371 PMCID: PMC5961502 DOI: 10.1038/nm.4424] [Citation(s) in RCA: 327] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 09/08/2017] [Indexed: 12/11/2022]
Abstract
De novo and acquired resistance, which are largely attributed to genetic alterations, are barriers to effective anti-epidermal-growth-factor-receptor (EGFR) therapy. To generate cetuximab-resistant cells, we exposed cetuximab-sensitive colorectal cancer cells to cetuximab in three-dimensional culture. Using whole-exome sequencing and transcriptional profiling, we found that the long non-coding RNA MIR100HG and two embedded microRNAs, miR-100 and miR-125b, were overexpressed in the absence of known genetic events linked to cetuximab resistance. MIR100HG, miR-100 and miR-125b overexpression was also observed in cetuximab-resistant colorectal cancer and head and neck squamous cell cancer cell lines and in tumors from colorectal cancer patients that progressed on cetuximab. miR-100 and miR-125b coordinately repressed five Wnt/β-catenin negative regulators, resulting in increased Wnt signaling, and Wnt inhibition in cetuximab-resistant cells restored cetuximab responsiveness. Our results describe a double-negative feedback loop between MIR100HG and the transcription factor GATA6, whereby GATA6 represses MIR100HG, but this repression is relieved by miR-125b targeting of GATA6. These findings identify a clinically actionable, epigenetic cause of cetuximab resistance.
Collapse
Affiliation(s)
- Yuanyuan Lu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xiaodi Zhao
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Qi Liu
- Department of Biomedical Informatics and Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cunxi Li
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China, and Molecular Pathology, Cancer Research Center, Medical College of Xiamen University, Xiamen, China
| | - Ramona Graves-Deal
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Zheng Cao
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bhuminder Singh
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeffrey L Franklin
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jing Wang
- Department of Biomedical Informatics and Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Huaying Hu
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China, and Molecular Pathology, Cancer Research Center, Medical College of Xiamen University, Xiamen, China
| | - Tianying Wei
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China, and Molecular Pathology, Cancer Research Center, Medical College of Xiamen University, Xiamen, China
| | - Mingli Yang
- Gibbs Cancer Center & Research Institute, Spartanburg, South Carolina, USA
| | - Timothy J Yeatman
- Gibbs Cancer Center & Research Institute, Spartanburg, South Carolina, USA
| | - Ethan Lee
- Department of Cell and Developmental Biology and Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kenyi Saito-Diaz
- Department of Cell and Developmental Biology and Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Scott Hinger
- Department of Biological Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - James G Patton
- Department of Biological Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | - Stephan Emmrich
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | | | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
163
|
Hartung N, Benary U, Wolf J, Kofahl B. Paracrine and autocrine regulation of gene expression by Wnt-inhibitor Dickkopf in wild-type and mutant hepatocytes. BMC SYSTEMS BIOLOGY 2017; 11:98. [PMID: 29029622 PMCID: PMC5640931 DOI: 10.1186/s12918-017-0470-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 10/02/2017] [Indexed: 12/12/2022]
Abstract
Background Cells are able to communicate and coordinate their function within tissues via secreted factors. Aberrant secretion by cancer cells can modulate this intercellular communication, in particular in highly organised tissues such as the liver. Hepatocytes, the major cell type of the liver, secrete Dickkopf (Dkk), which inhibits Wnt/ β-catenin signalling in an autocrine and paracrine manner. Consequently, Dkk modulates the expression of Wnt/ β-catenin target genes. We present a mathematical model that describes the autocrine and paracrine regulation of hepatic gene expression by Dkk under wild-type conditions as well as in the presence of mutant cells. Results Our spatial model describes the competition of Dkk and Wnt at receptor level, intra-cellular Wnt/ β-catenin signalling, and the regulation of target gene expression for 21 individual hepatocytes. Autocrine and paracrine regulation is mediated through a feedback mechanism via Dkk and Dkk diffusion along the porto-central axis. Along this axis an APC concentration gradient is modelled as experimentally detected in liver. Simulations of mutant cells demonstrate that already a single mutant cell increases overall Dkk concentration. The influence of the mutant cell on gene expression of surrounding wild-type hepatocytes is limited in magnitude and restricted to hepatocytes in close proximity. To explore the underlying molecular mechanisms, we perform a comprehensive analysis of the model parameters such as diffusion coefficient, mutation strength and feedback strength. Conclusions Our simulations show that Dkk concentration is elevated in the presence of a mutant cell. However, the impact of these elevated Dkk levels on wild-type hepatocytes is confined in space and magnitude. The combination of inter- and intracellular processes, such as Dkk feedback, diffusion and Wnt/ β-catenin signal transduction, allow wild-type hepatocytes to largely maintain their gene expression. Electronic supplementary material The online version of this article (doi:10.1186/s12918-017-0470-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Niklas Hartung
- University of Potsdam, Institute of Mathematics, Karl-Liebknecht-Str. 24, Potsdam, 14476, Germany
| | - Uwe Benary
- Mathematical Modelling of Cellular Processes, Max Delbrueck Center for Molecular Medicine, Robert-Roessle-Str. 10, Berlin, 13125, Germany
| | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max Delbrueck Center for Molecular Medicine, Robert-Roessle-Str. 10, Berlin, 13125, Germany
| | - Bente Kofahl
- Mathematical Modelling of Cellular Processes, Max Delbrueck Center for Molecular Medicine, Robert-Roessle-Str. 10, Berlin, 13125, Germany. .,Current address: Institute of Physics, University of Freiburg, Hermann-Herder-Str. 3, Freiburg i. Br., 79104, Germany.
| |
Collapse
|
164
|
Agley CC, Lewis FC, Jaka O, Lazarus NR, Velloso C, Francis-West P, Ellison-Hughes GM, Harridge SDR. Active GSK3β and an intact β-catenin TCF complex are essential for the differentiation of human myogenic progenitor cells. Sci Rep 2017; 7:13189. [PMID: 29030569 PMCID: PMC5640663 DOI: 10.1038/s41598-017-10731-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/14/2017] [Indexed: 01/14/2023] Open
Abstract
Wnt-β-catenin signalling is essential for skeletal muscle myogenesis during development, but its role in adult human skeletal muscle remains unknown. Here we have used human primary CD56Pos satellite cell-derived myogenic progenitors obtained from healthy individuals to study the role of Wnt-β-catenin signalling in myogenic differentiation. We show that dephosphorylated β-catenin (active-β-catenin), the central effector of the canonical Wnt cascade, is strongly upregulated at the onset of differentiation and undergoes nuclear translocation as differentiation progresses. To establish the role of Wnt signalling in regulating the differentiation process we manipulated key nodes of this pathway through a series of β-catenin gain-of-function (GSK3 inhibition and β-catenin overexpression) or loss-of-function experiments (dominant negative TCF4). Our data showed that manipulation of these critical pathway components led to varying degrees of disruption to the normal differentiation phenotype indicating the importance of Wnt signalling in regulating this process. We reveal an independent necessity for active-β-catenin in the fusion and differentiation of human myogenic progenitors and that dominant negative inhibition of TCF4 prevents differentiation completely. Together these data add new mechanistic insights into both Wnt signalling and adult human myogenic progenitor differentiation.
Collapse
Affiliation(s)
- C C Agley
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK. .,Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| | - F C Lewis
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK.,Stem Cell Institute, King's College London, London, UK
| | - O Jaka
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK
| | - N R Lazarus
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK
| | - C Velloso
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK
| | - P Francis-West
- Department of Craniofacial development and stem cell biology, King's College London, London, UK
| | - G M Ellison-Hughes
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK.,Stem Cell Institute, King's College London, London, UK
| | - S D R Harridge
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK.,Stem Cell Institute, King's College London, London, UK
| |
Collapse
|
165
|
Susman MW, Karuna EP, Kunz RC, Gujral TS, Cantú AV, Choi SS, Jong BY, Okada K, Scales MK, Hum J, Hu LS, Kirschner MW, Nishinakamura R, Yamada S, Laird DJ, Jao LE, Gygi SP, Greenberg ME, Ho HYH. Kinesin superfamily protein Kif26b links Wnt5a-Ror signaling to the control of cell and tissue behaviors in vertebrates. eLife 2017; 6:e26509. [PMID: 28885975 PMCID: PMC5590807 DOI: 10.7554/elife.26509] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022] Open
Abstract
Wnt5a-Ror signaling constitutes a developmental pathway crucial for embryonic tissue morphogenesis, reproduction and adult tissue regeneration, yet the molecular mechanisms by which the Wnt5a-Ror pathway mediates these processes are largely unknown. Using a proteomic screen, we identify the kinesin superfamily protein Kif26b as a downstream target of the Wnt5a-Ror pathway. Wnt5a-Ror, through a process independent of the canonical Wnt/β-catenin-dependent pathway, regulates the cellular stability of Kif26b by inducing its degradation via the ubiquitin-proteasome system. Through this mechanism, Kif26b modulates the migratory behavior of cultured mesenchymal cells in a Wnt5a-dependent manner. Genetic perturbation of Kif26b function in vivo caused embryonic axis malformations and depletion of primordial germ cells in the developing gonad, two phenotypes characteristic of disrupted Wnt5a-Ror signaling. These findings indicate that Kif26b links Wnt5a-Ror signaling to the control of morphogenetic cell and tissue behaviors in vertebrates and reveal a new role for regulated proteolysis in noncanonical Wnt5a-Ror signal transduction.
Collapse
Affiliation(s)
- Michael W Susman
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| | - Edith P Karuna
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Ryan C Kunz
- Department of Cell BiologyHarvard Medical SchoolBostonUnited States
| | - Taranjit S Gujral
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
- Division of Human BiologyFred Hutchinson Cancer Research CenterSeattleUnited States
| | - Andrea V Cantú
- Department of Obstetrics, Gynecology and Reproductive SciencesCenter for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of CaliforniaSan FranciscoUnited States
| | - Shannon S Choi
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Brigette Y Jong
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Kyoko Okada
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Michael K Scales
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Jennie Hum
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Linda S Hu
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| | - Marc W Kirschner
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
| | - Ryuichi Nishinakamura
- Department of Kidney DevelopmentInstitute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Soichiro Yamada
- Department of Biomedical EngineeringUniversity of CaliforniaDavisUnited States
| | - Diana J Laird
- Department of Obstetrics, Gynecology and Reproductive SciencesCenter for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of CaliforniaSan FranciscoUnited States
| | - Li-En Jao
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Steven P Gygi
- Department of Cell BiologyHarvard Medical SchoolBostonUnited States
| | | | - Hsin-Yi Henry Ho
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| |
Collapse
|
166
|
Chen EEM, Zhang W, Ye CCY, Gao X, Jiang LLJ, Zhao TTF, Pan ZZJ, Xue DDT. Knockdown of SIRT7 enhances the osteogenic differentiation of human bone marrow mesenchymal stem cells partly via activation of the Wnt/β-catenin signaling pathway. Cell Death Dis 2017; 8:e3042. [PMID: 28880264 PMCID: PMC5636975 DOI: 10.1038/cddis.2017.429] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 07/08/2017] [Accepted: 07/25/2017] [Indexed: 01/13/2023]
Abstract
Sirtuin 7 (SIRT7) is a NAD+-dependent deacetylase in the sirtuin family. In a previous study, human bone marrow mesenchymal stem cells (hBMSCs) with reduced SIRT7 activity were developed to evaluate the effect of SIRT7 on osteogenesis. SIRT7 knockdown significantly enhanced osteoblast-specific gene expression, alkaline phosphatase activity, and mineral deposition in vitro. Additionally, SIRT7 knockdown upregulated β-catenin. The enhanced osteogenesis due to SIRT7 knockdown was partially rescued by a Wnt/β-catenin inhibitor. Furthermore, SIRT7 knockdown hBMSCs combined with a chitosan scaffold significantly promoted bone formation in a rat tibial defect model, as determined by imaging and histological examinations. These findings suggest that SIRT7 has an essential role in osteogenic differentiation of hBMSCs, partly by activation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Erman E M Chen
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People’s Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, People’s Republic of China
| | - Wei Zhang
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People’s Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, People’s Republic of China
| | - Chenyi C Y Ye
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People’s Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, People’s Republic of China
| | - Xiang Gao
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People’s Republic of China
| | - Liangjun L J Jiang
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People’s Republic of China
| | - Tengfei T F Zhao
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People’s Republic of China
| | - Zhijun Z J Pan
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People’s Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, People’s Republic of China
| | - Deting D T Xue
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People’s Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, People’s Republic of China
| |
Collapse
|
167
|
Tortelote GG, Reis RR, de Almeida Mendes F, Abreu JG. Complexity of the Wnt/β‑catenin pathway: Searching for an activation model. Cell Signal 2017; 40:30-43. [PMID: 28844868 DOI: 10.1016/j.cellsig.2017.08.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/08/2017] [Accepted: 08/23/2017] [Indexed: 12/13/2022]
Abstract
Wnt signaling refers to a conserved signaling pathway, widely studied due to its roles in cellular communication, cell fate decisions, development and cancer. However, the exact mechanism underlying inhibition of the GSK phosphorylation towards β-catenin and activation of the pathway after biding of Wnt ligand to its cognate receptors at the plasma membrane remains unclear. Wnt target genes are widely spread over several animal phyla. They participate in a plethora of functions during the development of an organism, from axial specification, gastrulation and organogenesis all the way to regeneration and repair in adults. Temporal and spatial oncogenetic re-activation of Wnt signaling almost certainly leads to cancer. Wnt signaling components have been extensively studied as possible targets in anti-cancer therapies. In this review we will discuss one of the most intriguing questions in this field, that is how β-catenin, a major component in this pathway, escapes the destruction complex, gets stabilized in the cytosol and it is translocated to the nucleus where it acts as a co-transcription factor. Four major models have evolved during the past 20years. We dissected each of them along with current views and future perspectives on this pathway. This review will focus on the molecular mechanisms by which Wnt proteins modulate β-catenin cytoplasmic levels and the relevance of this pathway for the development and cancer.
Collapse
Affiliation(s)
- Giovane G Tortelote
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Renata R Reis
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio de Almeida Mendes
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose Garcia Abreu
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
168
|
Jiang C, Yu M, Xie X, Huang G, Peng Y, Ren D, Lin M, Liu B, Liu M, Wang W, Kuang M. miR-217 targeting DKK1 promotes cancer stem cell properties via activation of the Wnt signaling pathway in hepatocellular carcinoma. Oncol Rep 2017; 38:2351-2359. [PMID: 28849121 DOI: 10.3892/or.2017.5924] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 05/24/2017] [Indexed: 11/06/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies and exhibits heterogeneity in terms of clinical outcomes and biological activities. Emerging evidence has demonstrated that cancer stem cells (CSCs) play important roles in the tumorigenesis and progression of HCC. However, the molecular mechanisms underlying the stemness maintenance of CSCs remain largely unknown. In the present study, through real-time PCR, western blotting, luciferase assays, RNA immunoprecipitation, and in vitro and in vivo assays, we demonstrated that miR-217 expression was markedly increased in HCC tissues and cells. Overexpression of miR-217 promoted, while silencing miR-217 suppressed, the fraction of the side population and the expression of cancer stem cell factors in vitro and tumorigenicity in vivo in HCC cells. Our findings further demonstrated that miR-217 promoted the CSC-like phenotype via dickkopf-1 (DKK1) targeting, resulting in constitutive activation of Wnt signaling. Moreover, the stimulatory effects of miR-217 on stem cell properties and Wnt signaling were antagonized by the upregulation of DKK1 in miR-217-overexpressing cells. Conversely, the inhibitory effects of silencing miR-217 on stem cell properties and Wnt signaling were reversed by the downregulation of DKK1 in miR-217-downregulated cells. Therefore, our results indicate that miR-217 plays a vital role in the CSC-like phenotypes of HCC cells and may be used as a potential therapeutic target against HCC.
Collapse
Affiliation(s)
- Chunlin Jiang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Miao Yu
- Center for Private Medical Service and Healthcare, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaoyan Xie
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Guangliang Huang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yao Peng
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Dong Ren
- Department of Orthopaedic Surgery/Orthopaedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Manxia Lin
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Baoxian Liu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Ming Liu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wei Wang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Ming Kuang
- Division of Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
169
|
Hochman G, Halevi-Tobias K, Kogan Y, Agur Z. Extracellular inhibitors can attenuate tumorigenic Wnt pathway activity in adenomatous polyposis coli mutants: Predictions of a validated mathematical model. PLoS One 2017; 12:e0179888. [PMID: 28708837 PMCID: PMC5510801 DOI: 10.1371/journal.pone.0179888] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/06/2017] [Indexed: 12/21/2022] Open
Abstract
Background Despite considerable investigational efforts, no method to overcome the pathogenesis caused by loss of function (LoF) mutations in tumor suppressor genes has been successfully translated to the clinic. The most frequent LoF mutation in human cancers is Adenomatous polyposis coli (APC), causing aberrant activation of the Wnt pathway. In nearly all colon cancer tumors, the APC protein is truncated, but still retains partial binding abilities. Objective & methods Here, we tested the hypothesis that extracellular inhibitors of the Wnt pathway, although acting upstream of the APC mutation, can restore normal levels of pathway activity in colon cancer cells. To this end, we developed and simulated a mathematical model for the Wnt pathway in different APC mutants, with or without the effects of the extracellular inhibitors, Secreted Frizzled-Related Protein1 (sFRP1) and Dickhopf1 (Dkk1). We compared our model predictions to experimental data in the literature. Results Our model accurately predicts T-cell factor (TCF) activity in mutant cells that vary in APC mutation. Model simulations suggest that both sFRP1 and DKK1 can reduce TCF activity in APC1638N/1572T and Apcmin/min mutants, but restoration of normal activity levels is possible only in the former. When applied in combination, synergism between the two inhibitors can reduce their effective doses to one-fourth of the doses required under single inhibitor application. Overall, re-establishment of normal Wnt pathway activity is predicted for every APC mutant in whom TCF activity is increased by up to 11 fold. Conclusions Our work suggests that extracellular inhibitors can effectively restore normal Wnt pathway activity in APC-truncated cancer cells, even though these LoF mutations occur downstream of the inhibitory action. The insufficient activity of the truncated APC can be quantitatively balanced by the upstream intervention. This new concept of upstream intervention to control the effects of downstream mutations may be considered also for other partial LoF mutations in other signaling pathways.
Collapse
Affiliation(s)
- Gili Hochman
- Institute for Medical BioMathematics, Bene Ataroth, Israel
| | | | - Yuri Kogan
- Institute for Medical BioMathematics, Bene Ataroth, Israel
| | - Zvia Agur
- Institute for Medical BioMathematics, Bene Ataroth, Israel
- * E-mail:
| |
Collapse
|
170
|
Niessner H, Kosnopfel C, Sinnberg T, Beck D, Krieg K, Wanke I, Lasithiotakis K, Bonin M, Garbe C, Meier F. Combined activity of temozolomide and the mTOR inhibitor temsirolimus in metastatic melanoma involves DKK1. Exp Dermatol 2017; 26:598-606. [PMID: 28423208 DOI: 10.1111/exd.13372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2017] [Indexed: 02/03/2023]
Abstract
The BRAFV600E inhibitor vemurafenib achieves remarkable clinical responses in patients with BRAF-mutant melanoma, but its effects are limited by the onset of drug resistance. In the case of resistance, chemotherapy can still be applied as second line therapy. However, it yields low response rates and strategies are urgently needed to potentiate its effects. In a previous study, we showed that the inhibition of the PI3K-AKT-mTOR pathway significantly increases sensitivity of melanoma cells to chemotherapeutic drugs (J. Invest. Dermatol. 2009, 129, 1500). In this study, the combination of the mTOR inhibitor temsirolimus with the chemotherapeutic agent temozolomide significantly increases growth inhibition and apoptosis in melanoma cells compared to temsirolimus or temozolomide alone. The combination of temozolomide with temsirolimus is not only effective in established but also in newly isolated and vemurafenib-resistant metastatic melanoma cell lines. These effects are associated with the downregulation of the anti-apoptotic protein Mcl-1 and the upregulation of the Wnt antagonist Dickkopf homologue 1 (DKK1). Knock-down of DKK1 suppresses apoptosis induction by the combination of temsirolimus and temozolomide. These data suggest that the inhibition of the mTOR pathway increases sensitivity of melanoma cells towards temozolomide. Chemosensitisation is associated with enhanced expression of the Wnt antagonist DKK1.
Collapse
Affiliation(s)
- Heike Niessner
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Corinna Kosnopfel
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Tobias Sinnberg
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Daniela Beck
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Kathrin Krieg
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Ines Wanke
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | | | - Michael Bonin
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Friedegund Meier
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| |
Collapse
|
171
|
Cuesta S, Severin MJ, Batuecas J, Rosso SB, Pacchioni AM. Wnt/β-catenin pathway in the prefrontal cortex is required for cocaine-induced neuroadaptations. Addict Biol 2017; 22:933-945. [PMID: 26910786 DOI: 10.1111/adb.12377] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 12/21/2022]
Abstract
Behavioral sensitization is a progressive and enduring enhancement of the motor stimulant effects elicited by repeated administration of drugs of abuse. It can be divided into two distinct temporal and anatomical domains, termed initiation and expression, which are characterized by specific molecular and neurochemical changes. This study examines the role of the Wnt canonical pathway mediating the induction of cocaine sensitization. We found that β-catenin levels in the prefrontal cortex (PFC), amygdala (Amyg) and dorsal striatum (CPu) are decreased in animals that show sensitization. Accordingly, GSK3β activity levels are increased in the same areas. Moreover, β-catenin levels in nuclear fraction, mRNA expression of Axin2 and Wnt7b are decreased in the PFC of sensitized animals. Then, in order to demonstrate that changes in the PFC are crucial for initiation of sensitization, we either rescue β-catenin levels with a systemic treatment of a GSK3β inhibitor (Lithium Chloride) or inhibit Wnt/β-catenin pathway with an intracerebral infusion of Sulindac before each cocaine injection. As expected, rescuing β-catenin levels in the PFC as well as CPu and Amyg blocks cocaine-induced sensitization, while decreasing β-catenin levels exclusively in the PFC exacerbates it. Therefore, our results demonstrate a new role for the Wnt/β-catenin pathway as a required neuroadaptation in inducing behavioral sensitization.
Collapse
Affiliation(s)
- Santiago Cuesta
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET); Argentina
- Douglas Mental Health University Institute; Canada
- Área Toxicología, Departamento de Ciencias de los Alimentos y del Medioambiente, Facultad de Ciencias Bioquímicas y Farmacéuticas; Universidad Nacional de Rosario (U.N.R); Argentina
| | - Maria J. Severin
- Área Toxicología, Departamento de Ciencias de los Alimentos y del Medioambiente, Facultad de Ciencias Bioquímicas y Farmacéuticas; Universidad Nacional de Rosario (U.N.R); Argentina
| | - Jorgelina Batuecas
- Área Toxicología, Departamento de Ciencias de los Alimentos y del Medioambiente, Facultad de Ciencias Bioquímicas y Farmacéuticas; Universidad Nacional de Rosario (U.N.R); Argentina
| | - Silvana B. Rosso
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET); Argentina
- Área Toxicología, Departamento de Ciencias de los Alimentos y del Medioambiente, Facultad de Ciencias Bioquímicas y Farmacéuticas; Universidad Nacional de Rosario (U.N.R); Argentina
| | - Alejandra M. Pacchioni
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET); Argentina
- Área Toxicología, Departamento de Ciencias de los Alimentos y del Medioambiente, Facultad de Ciencias Bioquímicas y Farmacéuticas; Universidad Nacional de Rosario (U.N.R); Argentina
| |
Collapse
|
172
|
Abou Ziki MD, Mani A. Wnt signaling, a novel pathway regulating blood pressure? State of the art review. Atherosclerosis 2017; 262:171-178. [PMID: 28522145 PMCID: PMC5508596 DOI: 10.1016/j.atherosclerosis.2017.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/06/2017] [Accepted: 05/03/2017] [Indexed: 12/18/2022]
Abstract
Recent antihypertensive trials show conflicting results on blood pressure (BP) targets in patient populations with different metabolic profiles, with lowest benefit from tight BP control observed in patients with type 2 diabetes mellitus. This paradox could arise from the heterogeneity of study populations and underscores the importance of precision medicine initiatives towards understanding and treating hypertension. Wnt signaling pathways and genetic variations in its signaling peptides have been recently associated with metabolic syndrome, hypertension and diabetes, generating a breakthrough for advancement of precision medicine in the field of hypertension. We performed a review of PubMed for publications addressing the contributions of Wnt to BP regulation and hypertension. In addition, we performed a manual search of the reference lists for relevant articles, and included unpublished observations from our laboratory. There is emerging evidence for Wnt's role in BP regulation and its involvement in the pathogenesis of hypertension. Wnt signaling has pleiotropic effects on distinct pathways that involve vascular smooth muscle plasticity, and cardiac, renal, and neural physiology. Hypertension is a heterogeneous disease with unique molecular pathways regulating its response to therapy. Recognition of these pathways is a prerequisite to identify novel targets for drug development and personalizing medicine. A review of Wnt signaling reveals its emerging role in BP regulation and as a target for novel drug development that has the potential to transform the therapy of hypertension in specific populations.
Collapse
Affiliation(s)
- Maen D Abou Ziki
- Departments of Internal Medicine and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Arya Mani
- Departments of Internal Medicine and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
173
|
Kjolby RAS, Harland RM. Genome-wide identification of Wnt/β-catenin transcriptional targets during Xenopus gastrulation. Dev Biol 2017; 426:165-175. [PMID: 27091726 PMCID: PMC6288011 DOI: 10.1016/j.ydbio.2016.03.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/16/2016] [Accepted: 03/16/2016] [Indexed: 10/21/2022]
Abstract
The canonical Wnt/β-catenin signaling pathway plays multiple roles during Xenopus gastrulation, including posteriorization of the neural plate, patterning of the mesoderm, and induction of the neural crest. Wnt signaling stabilizes β-catenin, which then activates target genes. However, few targets of this signaling pathway that mediate early developmental processes are known. Here we sought to identify transcriptional targets of the Wnt/β-catenin signaling pathway using a genome-wide approach. We selected putative targets using the criteria of reduced expression upon zygotic Wnt knockdown, β-catenin binding within 50kb of the gene, and expression in tissues that receive Wnt signaling. Using these criteria, we found 21 novel direct transcriptional targets of Wnt/β-catenin signaling during gastrulation and in addition have identified putative regulatory elements for further characterization in future studies.
Collapse
Affiliation(s)
- Rachel A S Kjolby
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Richard M Harland
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
174
|
Sopko R, Mugford JW, Lehmann A, Shapiro RI, Rushe M, Kulkarni A, Worrall J, Amatucci J, Wen D, Pederson NE, Minesinger BK, Arndt JW, Pepinsky B. Engineering potent long-acting variants of the Wnt inhibitor DKK2. Protein Eng Des Sel 2017; 30:359-372. [PMID: 28180900 PMCID: PMC5425732 DOI: 10.1093/protein/gzx007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/19/2017] [Indexed: 11/15/2022] Open
Abstract
Wnt signaling pathways are required for a wide variety of biological processes ranging from embryonic development to tissue repair and regeneration. Dickkopf-2 (DKK2) is classically defined as a canonical Wnt inhibitor, though it may play a role in activating non-canonical Wnt pathways in the context of endothelial network formation after acute injury. Here we report the discovery of a fusion partner for a DKK2 polypeptide that significantly improves the expression, biochemical properties and pharmacokinetics (PK) of the DKK2 polypeptide. Specifically, human serum albumin (HSA) was identified as a highly effective fusion partner. Substitution of selected amino acid residues in DKK2 designed to decrease heparan sulfate binding by HSA-DKK2 variants, further improved the PK properties of the molecule in rodents. The HSA-DKK2 variants were monomeric, as thermally stable as wild type, and active as measured by their ability to bind to and prevent phosphorylation of the Wnt coreceptor LRP6. Our engineering efforts resulted in potent long-lived variants of the canonical Wnt inhibitor DKK2, applicable for Wnt pathway manipulation either by systematic delivery or focused administration at sites of tissue injury.
Collapse
Affiliation(s)
- Richelle Sopko
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Joshua W Mugford
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Andreas Lehmann
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Renée I Shapiro
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Mia Rushe
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Abhishek Kulkarni
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Joseph Worrall
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Joseph Amatucci
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Dingyi Wen
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Nels E Pederson
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | | | - Joseph W Arndt
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| | - Blake Pepinsky
- Department of Cell and Protein Sciences, Biogen, Cambridge, MA 02142, USA
| |
Collapse
|
175
|
Li B, Liu J, Zhao J, Ma JX, Jia HB, Zhang Y, Xing GS, Ma XL. LncRNA-H19 Modulates Wnt/β-catenin Signaling by Targeting Dkk4 in Hindlimb Unloaded Rat. Orthop Surg 2017; 9:319-327. [PMID: 28447380 DOI: 10.1111/os.12321] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 02/18/2017] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To investigate the biological functions of long noncoding RNA-H19 (H19) in the pathogenesis of disuse osteoporosis (DOP). METHODS Fifty-four male Sprague Dawley (SD) rats were randomly divided into three groups: baseline control (BC, 6), age-matched control (AC, 24), and hindlimb unloading (HLU, 24). The rats in the BC group were sacrificed at the beginning of the experiment, while the AC and HLU rats were sacrificed at different times (7, 14, 21 and 28 days after HLU). The DOP model was verified by micro-CT scan, and quantitative real-time polymerase chain reaction (qRT-PCR) was used to quantify the expression of osteogenic genes (OPG, RunX2 and OPG). Gene sequencing and bioinformatic analysis were performed to find H19 target genes and the associated signaling pathway, which were first verified on tissue samples. Further verification was performed by knocking down the H19 and related gene in rat osteoblast cell line (UMR106 cell). Then, the changes of associated signaling pathway and osteogenic function were examined to confirm the prediction of the bioinformatic analysis. RESULTS Micro-CT scans and quantitative real-time polymerase chain reaction (qRT-PCR) tests showed progressively deteriorated trabecular bone and decreased level of osteogenic genes in the metaphysis of distal femur during HLU, indicating the successful establishment of a DOP model. According to RNA sequencing, 1351 mRNA and 464 lncRNA were abnormally expressed in response to mechanical unloading, in which the H19 decreased 2.86 fold in HLU rats. There were 1426 mRNA predicted to be the target genes of H19, and KEGG pathway analysis suggested that Wnt signaling pathway (Wnt signaling) was the top pathway responsible for these target genes. In the Wnt-associated genes targeted by H19, 11 were differentially expressed between HLU and AC rats, among which Dkk4 increased 2.44 fold in HLU rats when compared to normal controls. These results of sequencing and bioinformatic analysis were confirmed by the low expression of H19, overexpression of Dkk4 and inhibited Wnt signaling observed in DOP rats. Subsequent in vitro cell assay further demonstrated that knockdown of H19 led to upregulation of Dkk4, and inhibition of Wnt signaling and osteogenic function in UMR106 cell. These effects can be greatly reversed after application of knocking down Dkk4. CONCLUSION Our findings demonstrated that low expression of H19, induced by mechanical unloading, leads to development of DOP through inhibition of Wnt signaling by promoting Dkk4 expression.
Collapse
Affiliation(s)
- Bing Li
- Department of Joint, Tianjin Hospital, Tianjin, China
| | - Jun Liu
- Department of Joint, Tianjin Hospital, Tianjin, China
| | - Jie Zhao
- Department of Biomechanics, Orthopaedic Research Institute, Tianjin Hospital, Tianjin, China
| | - Jian-Xiong Ma
- Department of Biomechanics, Orthopaedic Research Institute, Tianjin Hospital, Tianjin, China
| | - Hao-Bo Jia
- Department of Biomechanics, Orthopaedic Research Institute, Tianjin Hospital, Tianjin, China
| | - Yang Zhang
- Department of Biomechanics, Orthopaedic Research Institute, Tianjin Hospital, Tianjin, China
| | - Guo-Sheng Xing
- Department of Biomechanics, Orthopaedic Research Institute, Tianjin Hospital, Tianjin, China
| | - Xin-Long Ma
- Department of Biomechanics, Orthopaedic Research Institute, Tianjin Hospital, Tianjin, China
| |
Collapse
|
176
|
Computationally Design of Inhibitory Peptides Against Wnt Signaling Pathway: In Silico Insight on Complex of DKK1 and LRP6. Int J Pept Res Ther 2017. [DOI: 10.1007/s10989-017-9589-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
177
|
Wu J, Liu S, Meng H, Qu T, Fu S, Wang Z, Yang J, Jin D, Yu B. Neuropeptide Y enhances proliferation and prevents apoptosis in rat bone marrow stromal cells in association with activation of the Wnt/β-catenin pathway in vitro. Stem Cell Res 2017; 21:74-84. [PMID: 28411439 DOI: 10.1016/j.scr.2017.04.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 03/31/2017] [Accepted: 04/02/2017] [Indexed: 01/03/2023] Open
Abstract
Neuropeptide Y (NPY) exhibits a critical but poorly understood regulatory signaling function and has been shown to promote proliferation, vascularization and migration in several types of cells and tissues. However, little is known about the specific role of NPY in the proliferation and apoptosis of bone marrow stromal cells (also known as bone marrow-derived mesenchymal stem cells, BMSCs), which contain a subpopulation of multipotent skeletal stem cells. Based on BrdU incorporation tests, Cell Counting Kit-8, flow cytometry, quantitative polymerase chain reaction and western blotting, we showed that NPY significantly promoted the proliferation of BMSCs in a concentration-dependent manner, with a maximal effect observed at a concentration of 10-10M for pro-proliferative and 10-12M for anti-apoptotic activities. Furthermore, NPY significantly increased the percentage of cells in S and G2/M phases. In addition, NPY exhibited a protective effect after 24h of serum starvation as illustrated by a reduction in the apoptosis rate, degree of nuclear condensation, and expression of apoptosis markers, including caspase-3, caspase-9 and Bax mRNA expression. NPY also increased the mRNA and protein expression levels of canonical Wnt signaling pathway proteins, including β-catenin and c-myc, during the induced proliferative and anti-apoptotic processes. However, the proliferative and anti-apoptotic activities of NPY were partially blocked by both PD160170 (1μM) and DKK1 (0.2μg/mL). These compounds also blocked the mRNA and protein expression of β-catenin, p-GSK-3β and c-myc. Therefore, the results of the present study demonstrated that NPY exerts a proliferative and protective effect on BMSCs in a dose- and time-dependent manner in vitro, and importantly, these effects may be mediated via its Y1 receptor and involved in activation of the canonical Wnt signaling pathway.
Collapse
Affiliation(s)
- Jianqun Wu
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Song Liu
- Department of Orthopedics, The Third Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province 510515, China; Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Huan Meng
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Tianyu Qu
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Su Fu
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Zhao Wang
- Department of Orthopedics, The Third Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province 510515, China; Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Jianguo Yang
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; Department of Orthopaedics, The First Hospital Huhhot, Huhhot, Inner Mongolia 010020, China
| | - Dan Jin
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Bin Yu
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
178
|
ISLET1-Dependent β-Catenin/Hedgehog Signaling Is Required for Outgrowth of the Lower Jaw. Mol Cell Biol 2017; 37:MCB.00590-16. [PMID: 28069742 DOI: 10.1128/mcb.00590-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 01/04/2017] [Indexed: 12/30/2022] Open
Abstract
Mandibular patterning information initially resides in the epithelium during development. However, how transcriptional regulation of epithelium-derived signaling controls morphogenesis of the mandible remains elusive. Using ShhCre to target the mandibular epithelium, we ablated transcription factor Islet1, resulting in a distally truncated mandible via unbalanced cell apoptosis and decreased cell proliferation in the distal mesenchyme. Loss of Islet1 caused a lack of cartilage at the distal tip, leading the fusion of two growing mandibular elements surrounding the rostral process of Meckel's cartilage. Loss of Islet1 results in dysregulation of mesenchymal genes important for morphogenesis of the mandibular arch. We revealed that Islet1 is required for the activation of epithelial β-catenin signaling via repression of Wnt antagonists. Reactivation of β-catenin in the epithelium of the Islet1 mutant rescued mandibular morphogenesis through sonic hedgehog (SHH) signaling to the mesenchyme. Furthermore, overexpression of a transgenic hedgehog ligand in the epithelium also partially restored outgrowth of the mandible. These data reveal functional roles for an ISLET1-dependent network integrating β-catenin/SHH signals in mesenchymal cell survival and outgrowth of the mandible during development.
Collapse
|
179
|
Tanaka S, Hosokawa H, Weinberg ES, Maegawa S. Chordin and dickkopf-1b are essential for the formation of head structures through activation of the FGF signaling pathway in zebrafish. Dev Biol 2017; 424:189-197. [PMID: 28259755 DOI: 10.1016/j.ydbio.2017.02.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 02/16/2017] [Accepted: 02/28/2017] [Indexed: 01/16/2023]
Abstract
The ability of the Spemann organizer to induce dorsal axis formation is dependent on downstream factors of the maternal Wnt/β-catenin signaling pathway. The fibroblast growth factor (FGF) signaling pathway has been identified as one of the downstream components of the maternal Wnt/β-catenin signaling pathway. The ability of the FGF signaling pathway to induce the formation of a dorsal axis with a complete head structure requires chordin (chd) expression; however, the molecular mechanisms involved in this developmental process, due to activation of FGF signaling, remain unclear. In this study, we showed that activation of the FGF signaling pathway induced the formation of complete head structures through the expression of chd and dickkopf-1b (dkk1b). Using the organizer-deficient maternal mutant, ichabod, we identified dkk1b as a novel downstream factor in the FGF signaling pathway. We also demonstrate that dkk1b expression is necessary, after activation of the FGF signaling pathway, to induce neuroectoderm patterning along the anteroposterior (AP) axis and for formation of complete head structures. Co-injection of chd and dkk1b mRNA resulted in the formation of a dorsal axis with a complete head structure in ichabod embryos, confirming the role of these factors in this developmental process. Unexpectedly, we found that chd induced dkk1b expression in ichabod embryos at the shield stage. However, chd failed to maintain dkk1b expression levels in cells of the shield and, subsequently, in the cells of the prechordal plate after mid-gastrula stage. In contrast, activation of the FGF signaling pathway maintained the dkk1b expression from the beginning of gastrulation to early somitogenesis. In conclusion, activation of the FGF signaling pathway induces the formation of a dorsal axis with a complete head structure through the expression of chd and subsequent maintenance of dkk1b expression levels.
Collapse
Affiliation(s)
- Shingo Tanaka
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Hiroshi Hosokawa
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto 606-8501, Japan
| | - Eric S Weinberg
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shingo Maegawa
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
180
|
Khalili S, Rasaee MJ, Bamdad T. 3D structure of DKK1 indicates its involvement in both canonical and non-canonical Wnt pathways. Mol Biol 2017. [DOI: 10.1134/s0026893317010095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
181
|
Elhaj Mahmoud D, Sassi N, Drissi G, Barsaoui M, Zitouna K, Sahli H, Kallel-Sellami M, Kanoun L, Cheour E, Laadhar L. sFRP3 and DKK1 Regulate Fibroblast-Like Synoviocytes Markers and Wnt Elements Expression Depending on Cellular Context. Immunol Invest 2017; 46:314-328. [PMID: 28151034 DOI: 10.1080/08820139.2016.1267204] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
CONTEXT Fibroblast-like synoviocytes (FLS) from rheumatoid arthritis (RA) display pathogenic behavior. Various members of the Wnt pathway, especially the canonical Wnt/β-catenin cascade, may contribute to autonomous RA FLS activation. It has been shown that the two Wnt inhibitors: sFRP3 and DKK1 contribute to several critical aspects of joint biology. However, their effects on RA FLS are poorly characterized. The aim of our study was to investigate the effects of sFRP3 and DKK1 on FLS markers, Wnt components, and target oncogenes expression by RA FLS and compare the findings to osteoarthritic (OA) FLS. MATERIALS AND METHODS RA and OA FLS were treated with sFRP3 and DKK1 for 6 days. Wnt signaling components (Wnt5a, LRP5 and β-catenin), Wnt target oncogenes (cyclin E1 and WISP1), and FLS markers (fibronectin and MMP3) were analyzed using western blotting and/or qRT-PCR. RESULTS Our data indicated that sFRP3 down-regulated the key gene β-catenin in RA FLS. sFRP3 decreased fibronectin, a well-known downstream effectors gene of Wnt/β-catenin pathway, and LRP5 expression in both RA and OA FLS. In OA FLS, sFRP3 induced increased expression of Wnt5a and MMP3 but did not affect their levels in RA FLS. On the other hand, DKK1 increased fibronectin expression in RA FLS and decreased its expression in OA FLS. CONCLUSION Our results confirm the involvement of Wnt signaling in FLS transformation and show that two inhibitors of the same cascade can regulate differently the same elements and that a single inhibitor can initiate signaling depending on cellular context. ABBREVIATIONS FLS: fibroblast-like synoviocytes; RA: rheumatoid arthritis; Wnt: Wingless; Fz: frizzled; LRP: Fz/low-density lipoprotein receptor protein; WISP1: Wnt1 inducible signaling pathway protein 1; sFRP: secreted Fz-related proteins; DKK: Dickkopf; OA: osteoarthritis; DMEM: Dulbecco's modified Eagle's medium; FBS: fetal bovine serum; PBS: phosphate buffered saline; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; ECL: enhanced chemiluminescence detection solution; MMP3: metaloproteinase 3; qRT-PCR: quantitative real-time polymerase chain reaction; S.D: standard deviation; CRD: cysteine-rich domain; MeCP2: methyl-CpG-binding protein; RANKL: nuclear factor-kappa B ligand.
Collapse
Affiliation(s)
- Dorra Elhaj Mahmoud
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia
| | - Nadia Sassi
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia
| | - Ghassen Drissi
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia.,b Department of Orthopedic Surgery and Traumatology , La Rabta Hospital , Tunis , Tunisia
| | - Maher Barsaoui
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia.,b Department of Orthopedic Surgery and Traumatology , La Rabta Hospital , Tunis , Tunisia
| | - Khaled Zitouna
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia.,b Department of Orthopedic Surgery and Traumatology , La Rabta Hospital , Tunis , Tunisia
| | - Hela Sahli
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia
| | - Maryam Kallel-Sellami
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia
| | - Lassad Kanoun
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia.,b Department of Orthopedic Surgery and Traumatology , La Rabta Hospital , Tunis , Tunisia
| | - Elhem Cheour
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia
| | - Lilia Laadhar
- a Immuno-Rheumatology Research laboratory, Rheumatology Department , La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia
| |
Collapse
|
182
|
Morphine administration induces change in anxiety-related behavior via Wnt/β-catenin signaling. Neurosci Lett 2017; 639:199-206. [DOI: 10.1016/j.neulet.2017.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/26/2016] [Accepted: 01/03/2017] [Indexed: 02/01/2023]
|
183
|
Gavriatopoulou M, Dimopoulos MA, Kastritis E, Terpos E. Emerging treatment approaches for myeloma-related bone disease. Expert Rev Hematol 2017; 10:217-228. [PMID: 28092987 DOI: 10.1080/17474086.2017.1283213] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Multiple myeloma is characterized by the presence of osteolytic lesions that leads to devastating skeletal-related events in the majority of patients. Myeloma bone disease is attributed to increased osteoclastic and suppressed osteoblastic activity. Areas covered: Bisphosphonates remain the main treatment option, however they have limitations on their own. Understanding the pathogenesis of myeloma bone disease may provide a roadmap for new therapeutic approaches. The pathway of RANKRANKLOPG pathway has revealed denosumab, a monoclonal antibody targeting RANKL as a novel emerging therapy for myeloma-related bone disease. Furthermore, the Wnt signaling inhibitors dicckopf-1 and sclerostin that are implicated in the pathogenesis of bone destruction of myeloma are now targeted by novel monoclonal antibodies. Activin-A is a TGF-beta superfamily member which increases osteoclast activity and inhibits osteoblast function in myeloma; sotatercept and other molecules targeting activin-A have entered into clinical development. Several other molecules and pathways that play an important role in the pathogenesis of bone destruction in myeloma, such as periostin, adiponectin, Notch and BTK signaling are also targeted in an attempt to develop novel therapies for myeloma-related bone disease. Expert commentary: We summarize the current advances in the biology of myeloma bone disease and the potential therapeutic targets.
Collapse
Affiliation(s)
- Maria Gavriatopoulou
- a Department of Clinical Therapeutics , National and Kapodistrian University of Athens School of Medicine , Athens , Greece
| | - Meletios A Dimopoulos
- a Department of Clinical Therapeutics , National and Kapodistrian University of Athens School of Medicine , Athens , Greece
| | - Efstathios Kastritis
- a Department of Clinical Therapeutics , National and Kapodistrian University of Athens School of Medicine , Athens , Greece
| | - Evangelos Terpos
- a Department of Clinical Therapeutics , National and Kapodistrian University of Athens School of Medicine , Athens , Greece
| |
Collapse
|
184
|
Jang J, Jung Y, Kim Y, Jho EH, Yoon Y. LPS-induced inflammatory response is suppressed by Wnt inhibitors, Dickkopf-1 and LGK974. Sci Rep 2017; 7:41612. [PMID: 28128299 PMCID: PMC5269682 DOI: 10.1038/srep41612] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 12/21/2016] [Indexed: 12/24/2022] Open
Abstract
In this study, LPS-induced inflammatory responses in BEAS-2B human bronchial epithelial cells and human umbilical vein endothelial cell (HUVEC)s were found to be prevented by Dickkopf-1 (DKK-1), a secreted Wnt antagonist, and LGK974, a small molecular inhibitor of the Wnt secretion. LPS-induced IκB degradation and NF-κB nuclear translocation as well as the expressions of pro-inflammatory genes including IL-6, IL-8, TNF- α, IL-1β, MCP-1, MMP-9, COX-2 and iNOS, were all suppressed by DKK-1 and LGK974 in a dose-dependent manner. The suppressive effects of LGK974 on NF-κB, IκB, and pro-inflammatory gene expression were rescued by ectopic expression of β-catenin, suggesting that the anti-inflammatory activity of LGK974 is mediated by modulation of the Wnt/β-catenin pathway and not by unrelated side effects. When Wnt recombinant proteins were treated to cells, Wnt3a and Wnt5a significantly induced pro-inflammatory gene expressions, while Wnt7a and Wnt10b showed little effects. It was also found that Wnt3a and Wnt5a expressions were significantly induced by LPS treatment. Consistently, knockdown of Wnt3a and Wnt5a blocked LPS-induced inflammatory responses, while treatment of recombinant Wnt3a and Wnt5a proteins rescued the inhibition of inflammatory responses by LGK974. Findings of this study showed that DKK-1 and LGK974 suppress LPS-induced inflammatory response by modulating Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jaewoong Jang
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul 156-756, Republic of Korea
| | - Yoonju Jung
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul 156-756, Republic of Korea
| | - Youngeun Kim
- Department of Life Science, University of Seoul, Seoul, 130-743, Republic of Korea
| | - Eek-Hoon Jho
- Department of Life Science, University of Seoul, Seoul, 130-743, Republic of Korea
| | - Yoosik Yoon
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul 156-756, Republic of Korea
| |
Collapse
|
185
|
Rosenbloom J, Macarak E, Piera-Velazquez S, Jimenez SA. Human Fibrotic Diseases: Current Challenges in Fibrosis Research. Methods Mol Biol 2017; 1627:1-23. [PMID: 28836191 DOI: 10.1007/978-1-4939-7113-8_1] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Human fibrotic diseases constitute a major health problem worldwide owing to the large number of affected individuals, the incomplete knowledge of the fibrotic process pathogenesis, the marked heterogeneity in their etiology and clinical manifestations, the absence of appropriate and fully validated biomarkers, and, most importantly, the current void of effective disease-modifying therapeutic agents. The fibrotic disorders encompass a wide spectrum of clinical entities including systemic fibrotic diseases such as systemic sclerosis (SSc), sclerodermatous graft vs. host disease, and nephrogenic systemic fibrosis, as well as numerous organ-specific disorders including radiation-induced fibrosis and cardiac, pulmonary, liver, and kidney fibrosis. Although their causative mechanisms are quite diverse and in several instances have remained elusive, these diseases share the common feature of an uncontrolled and progressive accumulation of fibrotic tissue in affected organs causing their dysfunction and ultimate failure. Despite the remarkable heterogeneity in the etiologic mechanisms responsible for the development of fibrotic diseases and in their clinical manifestations, numerous studies have identified activated myofibroblasts as the common cellular element ultimately responsible for the replacement of normal tissues with nonfunctional fibrotic tissue. Critical signaling cascades, initiated primarily by transforming growth factor-β (TGF-β), but also involving numerous cytokines and signaling molecules which stimulate profibrotic reactions in myofibroblasts, offer potential therapeutic targets. Here, we briefly review the current knowledge of the molecular mechanisms involved in the development of tissue fibrosis and point out some of the most important challenges to research in the fibrotic diseases and to the development of effective therapeutic approaches for this often fatal group of disorders. Efforts to further clarify the complex pathogenetic mechanisms of the fibrotic process should be encouraged to attain the elusive goal of developing effective therapies for these serious, untreatable, and often fatal disorders.
Collapse
Affiliation(s)
- Joel Rosenbloom
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases and The Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Edward Macarak
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases and The Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sonsoles Piera-Velazquez
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases and The Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sergio A Jimenez
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases and The Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
186
|
Suttamanatwong S. MicroRNAs in bone development and their diagnostic and therapeutic potentials in osteoporosis. Connect Tissue Res 2017; 58:90-102. [PMID: 26963177 DOI: 10.3109/03008207.2016.1139580] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs approximately 22 nucleotides in length. miRNAs play an important role in the posttranscriptional regulation of gene expression via translational repression and targeting messenger RNA for degradation. In vivo and in vitro evidence has established the importance of miRNAs in physiology and developmental processes such as cell proliferation, differentiation, survival and apoptosis. miRNA dysregulation is associated with the pathogenesis of cardiovascular diseases, metabolic syndromes, and degenerative diseases. An increasing number of miRNAs have been found to play an important role in bone homeostasis. In this review, the roles of miRNAs in the regulation of bone formation and resorption as well as miRNAs that regulate key transcription factors of osteogenesis are discussed. A special emphasis is given to miRNAs whose direct targets have been identified. The miRNAs that contribute to the pathogenesis of osteoporosis and their therapeutic potential are also considered.
Collapse
Affiliation(s)
- Supaporn Suttamanatwong
- a Research Unit of Herbal Medicine, Biomaterial and Material for Dental Treatment, Department of Physiology, Faculty of Dentistry , Chulalongkorn University , Bangkok , Thailand
| |
Collapse
|
187
|
|
188
|
Tanaka M, Kuriyama S, Itoh G, Maeda D, Goto A, Tamiya Y, Yanagihara K, Yashiro M, Aiba N. Mesothelial Cells Create a Novel Tissue Niche That Facilitates Gastric Cancer Invasion. Cancer Res 2016; 77:684-695. [PMID: 27895076 DOI: 10.1158/0008-5472.can-16-0964] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 10/28/2016] [Accepted: 11/01/2016] [Indexed: 11/16/2022]
Abstract
Peritoneal mesothelial cells (PMC) cover organ surfaces in the abdominal cavity. In this study, lineage tracing revealed that the PMCs guide cancer cell invasion in the gastric wall and in peritoneal metastatic lesions. Serosal PMCs covering the stomach surface entered the gastric wall to create a novel niche that favored gastric cancer cell invasion. PMC infiltration was induced by incorporation of cancer cell-derived, Wnt3a-containing extracellular vesicles. Infiltrated PMCs in turn promoted subserosal invasion of cancer cells. Mutual attraction between cancer cells and PMCs accelerated tumor invasion in the gastric wall, and PMC-led cancer cell invasion in disseminated tumors within the abdominal wall and diaphragm. Addition of the carboxyl terminus of Dickkopf-1 attenuated directional invasion of PMCs toward cancer cells both in vitro and in the gastric wall in vivo PMCs were sensitive to the aldehyde dehydrogenase (ALDH) inhibitor disulfiram (DSF), as ALDH activity is elevated in PMCs. Wnt3a upregulated ALDH, and addition of DSF inhibited the invasive properties of PMCs, whereas DSF pretreatment suppressed gastric infiltration of PMCs and subserosal invasion by cancer cells. Our results suggest that stabilization of PMCs may become an effective therapy for the prevention of local invasion and metastasis of gastric cancer. Cancer Res; 77(3); 684-95. ©2016 AACR.
Collapse
Affiliation(s)
- Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan.
| | - Sei Kuriyama
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Go Itoh
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Daichi Maeda
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Akiteru Goto
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yutaro Tamiya
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan.,Department of Life Science, Faculty and Graduate School of Engineering and Resource Science, Akita University, Akita, Japan
| | - Kazuyoshi Yanagihara
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Namiko Aiba
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
189
|
Inoue J, Fujita H, Bando T, Kondo Y, Kumon H, Ohuchi H. Expression analysis of Dickkopf-related protein 3 (Dkk3) suggests its pleiotropic roles for a secretory glycoprotein in adult mouse. J Mol Histol 2016; 48:29-39. [DOI: 10.1007/s10735-016-9703-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/08/2016] [Indexed: 12/18/2022]
|
190
|
Sima J, Piao Y, Chen Y, Schlessinger D. Molecular dynamics of Dkk4 modulates Wnt action and regulates meibomian gland development. Development 2016; 143:4723-4735. [PMID: 27864382 DOI: 10.1242/dev.143909] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/06/2016] [Indexed: 01/04/2023]
Abstract
Secreted Dickkopf (Dkk) proteins are major Wnt pathway modulators during organ development. Dkk1 has been widely studied and acts as a general Wnt inhibitor. However, the molecular function of other Dkks remains largely unknown. Here, we show that Dkk4 selectively inhibits a subset of Wnts, but is further inactivated by proteolytic cleavage. Meibomian gland (MG) formation is employed as a model where Dkk4 and its Wnt targets are expressed. Skin-specific expression of Dkk4 arrests MG growth at early germ phase, which is similar to that observed in Eda-ablated Tabby mice. Consistent with transient Dkk4 action, intact Dkk4 inhibits MG extension but the cleaved form progressively increases during MG development with a concomitant upswing in Wnt activity. Furthermore, both Dkk4 and its receptor (and Wnt co-receptor) Lrp6 are direct Eda targets during MG induction. In cell and organotypic cultures, Dkk4 inhibition is eliminated by elevation of Lrp6. Also, Lrp6 upregulation restores MG formation in Tabby mice. Thus, the dynamic state of Dkk4 itself and its interaction with Lrp6 modulates Wnt function during MG development, with a novel limitation of Dkk4 action by proteolytic cleavage.
Collapse
Affiliation(s)
- Jian Sima
- Laboratory of Genetics and Genomics, NIA/NIH-IRP, 251 Bayview Blvd, room 10B014, Baltimore, MD 21224, USA
| | - Yulan Piao
- Laboratory of Genetics and Genomics, NIA/NIH-IRP, 251 Bayview Blvd, room 10B014, Baltimore, MD 21224, USA
| | - Yaohui Chen
- Laboratory of Genetics and Genomics, NIA/NIH-IRP, 251 Bayview Blvd, room 10B014, Baltimore, MD 21224, USA
| | - David Schlessinger
- Laboratory of Genetics and Genomics, NIA/NIH-IRP, 251 Bayview Blvd, room 10B014, Baltimore, MD 21224, USA
| |
Collapse
|
191
|
Nickho H, Younesi V, Aghebati-Maleki L, Motallebnezhad M, Majidi Zolbanin J, Movassagh Pour A, Yousefi M. Developing and characterization of single chain variable fragment (scFv) antibody against frizzled 7 (Fzd7) receptor. Bioengineered 2016; 8:501-510. [PMID: 27849134 DOI: 10.1080/21655979.2016.1255383] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
ABSTACT Wnt/β-catenin signaling pathway through Frizzled receptors has been shown to play a key role in both normal development and tumorigenesis. Overexpression of Wnt pathway genes, such as Fzd7 in several malignancies is well-documented. Therefore, targeting of Fzd7 and its ligand inhibits cancer cells proliferation metastasis. In the present study we isolated single chain variable fragments (scFvs) against Fzd7 receptor using phage display method. Semi-synthetic human naive antibody libraries (Tomlinson I + J) was employed in panning procedure to isolate specific scFv against specific peptide from extracellular domain of Fzd7 receptor. The reactivity and growth inhibition effects of the selected antibodies was evaluated using enzyme-linked immunosorbent assay (ELISA), MTT and annexin V assays, respectively. Seven scFvs reactive to Fzd7 were selected following 4 rounds of panning. The results showed that the selected scFvs inhibits cell growth through apoptosis cell death in a triple negative breast cancer cells, MDA-MB-231. Given that Fzd7 and Wnt pathway plays a critical role in tumor progression, selected blocking scFvs represent significant potential for immunotherapy of breast cancer cells.
Collapse
Affiliation(s)
- Hamid Nickho
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Vahid Younesi
- d Pishtaz Teb Zaman Diagnostics , Tehran , Iran.,e Department of Laboratory Sciences , Faculty of Paramedical Sciences, Alborz University of Medical Sciences , Karaj , Iran
| | - Leili Aghebati-Maleki
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Morteza Motallebnezhad
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Jafar Majidi Zolbanin
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Aliakbar Movassagh Pour
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Mehdi Yousefi
- b Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
192
|
Wang X, Jia Y, Fei C, Song X, Li L. Activation/Proliferation-associated Protein 2 (Caprin-2) Positively Regulates CDK14/Cyclin Y-mediated Lipoprotein Receptor-related Protein 5 and 6 (LRP5/6) Constitutive Phosphorylation. J Biol Chem 2016; 291:26427-26434. [PMID: 27821587 DOI: 10.1074/jbc.m116.744607] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/03/2016] [Indexed: 01/13/2023] Open
Abstract
Low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6) are co-receptors for Wnt ligands. Upon ligand binding, LRP5/6 undergo glycogen synthase kinase 3 (GSK3)/casein kinase I (CKI)-mediated phosphorylation at multiple PPP(S/T)P motifs in the intracellular domain, which is essential for canonical Wnt signal transduction. On the other hand, in the Wnt-off state, the mitosis-specific CDK14-Cyclin Y kinase complex phosphorylates Ser-1490 of LRP5/6 at G2/M, thereby priming the receptor for Wnt-induced phosphorylation. However, it remains unclear how CDK14/Cyclin Y is recruited to LRP5/6 and whether there are other cofactors involved in this process. Previously, we identified Caprin-2 as a positive regulator of canonical Wnt signaling by promoting GSK3-depedent LRP5/6 phosphorylation upon Wnt stimulation. Here we uncovered that Caprin-2 positively regulates constitutive LRP5/6 Ser-1490 phosphorylation by complexing with CDK14/Cyclin Y. Caprin-2-mediated LRP5/6 phosphorylation is cell cycle-dependent in a pattern similar to that of CDK14/Cyclin Y-dependent LRP5/6 phosphorylation. Moreover, knockdown of Caprin-2 disrupts not only the interaction between CDK14 and Cyclin Y but also the interaction between CDK14/Cyclin Y and LRP6. Overall, our findings revealed an unrecognized role of Caprin-2 in facilitating LRP5/6 constitutive phosphorylation at G2/M through forming a quaternary complex with CDK14, Cyclin Y, and LRP5/6.
Collapse
Affiliation(s)
- Xin Wang
- From the State Key Laboratory of Molecular Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yingying Jia
- From the State Key Laboratory of Molecular Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cong Fei
- From the State Key Laboratory of Molecular Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaomin Song
- From the State Key Laboratory of Molecular Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lin Li
- From the State Key Laboratory of Molecular Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
193
|
Xiong Z, Guo M, Yu Y, Zhang FF, Ge MK, Chen GQ, Shen SM. Downregulation of AIF by HIF-1 contributes to hypoxia-induced epithelial–mesenchymal transition of colon cancer. Carcinogenesis 2016; 37:1079-1088. [DOI: 10.1093/carcin/bgw089] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
194
|
Mori Y, Adams D, Hagiwara Y, Yoshida R, Kamimura M, Itoi E, Rowe DW. Identification of a progenitor cell population destined to form fracture fibrocartilage callus in Dickkopf-related protein 3-green fluorescent protein reporter mice. J Bone Miner Metab 2016; 34:606-614. [PMID: 26369320 DOI: 10.1007/s00774-015-0711-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/02/2015] [Indexed: 01/23/2023]
Abstract
Fracture healing is a complex biological process involving the proliferation of mesenchymal progenitor cells, and chondrogenic, osteogenic, and angiogenic differentiation. The mechanisms underlying the proliferation and differentiation of mesenchymal progenitor cells remain unclear. Here, we demonstrate Dickkopf-related protein 3 (Dkk3) expression in periosteal cells using Dkk3-green fluorescent protein reporter mice. We found that proliferation of mesenchymal progenitor cells began in the periosteum, involving Dkk3-positive cell proliferation near the fracture site. In addition, Dkk3 was expressed in fibrocartilage cells together with smooth muscle α-actin and Col3.6 in the early phase of fracture healing as a cell marker of fibrocartilage cells. Dkk3 was not expressed in mature chondrogenic cells or osteogenic cells. Transient expression of Dkk3 disappeared in the late phase of fracture healing, except in the superficial periosteal area of fracture callus. The Dkk3 expression pattern differed in newly formed type IV collagen positive blood vessels and the related avascular tissue. This is the first report that shows Dkk3 expression in the periosteum at a resting state and in fibrocartilage cells during the fracture healing process, which was associated with smooth muscle α-actin and Col3.6 expression in mesenchymal progenitor cells. These fluorescent mesenchymal lineage cells may be useful for future studies to better understand fracture healing.
Collapse
Affiliation(s)
- Yu Mori
- Center for Regenerative Medicine and Skeletal Biology, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA.
- Department of Orthopaedic Surgery, Graduate School of Tohoku University, 1-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8574, Japan.
| | - Douglas Adams
- Center for Regenerative Medicine and Skeletal Biology, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Yusuke Hagiwara
- Center for Regenerative Medicine and Skeletal Biology, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Ryu Yoshida
- Center for Regenerative Medicine and Skeletal Biology, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Masayuki Kamimura
- Department of Orthopaedic Surgery, Graduate School of Tohoku University, 1-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8574, Japan
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Graduate School of Tohoku University, 1-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8574, Japan
| | - David W Rowe
- Center for Regenerative Medicine and Skeletal Biology, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
195
|
Zhong L, Huang X, Rodrigues ED, Leijten JCH, Verrips T, El Khattabi M, Karperien M, Post JN. Endogenous DKK1 and FRZB Regulate Chondrogenesis and Hypertrophy in Three-Dimensional Cultures of Human Chondrocytes and Human Mesenchymal Stem Cells. Stem Cells Dev 2016; 25:1808-1817. [PMID: 27733096 PMCID: PMC5124737 DOI: 10.1089/scd.2016.0222] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hypertrophic differentiation occurs during in vitro chondrogenesis of mesenchymal stem cells (MSCs), decreasing the quality of the cartilage construct. Previously we identified WNT pathway antagonists Dickkopf 1 homolog (DKK1) and frizzled-related protein (FRZB) as key factors in blocking hypertrophic differentiation of human MSCs (hMSCs). In this study, we investigated the role of endogenously expressed DKK1 and FRZB in chondrogenesis of hMSC and chondrocyte redifferentiation and in preventing cell hypertrophy using three relevant human cell based systems, isolated hMSCs, isolated primary human chondrocytes (hChs), and cocultures of hMSCs with hChs for which we specifically designed neutralizing nano-antibodies. We selected and tested variable domain of single chain heavy chain only antibodies (VHH) for their ability to neutralize the function of DKK1 or FRZB. In the presence of DKK1 and FRZB neutralizing VHH, glycosaminoglycan and collagen type II staining were significantly reduced in monocultured hMSCs and monocultured chondrocytes. Furthermore, in cocultures, cells in pellets showed hypertrophic differentiation. In conclusion, endogenous expression of the WNT antagonists DKK1 and FRZB is necessary for multiple steps during chondrogenesis: first DKK1 and FRZB are indispensable for the initial steps of chondrogenic differentiation of hMSCs, second they are necessary for chondrocyte redifferentiation, and finally in preventing hypertrophic differentiation of articular chondrocytes.
Collapse
Affiliation(s)
- Leilei Zhong
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Xiaobin Huang
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Emilie Dooms Rodrigues
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Jeroen C H Leijten
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | | | | | - Marcel Karperien
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Janine N Post
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| |
Collapse
|
196
|
Bhavanasi D, Klein PS. Wnt Signaling in Normal and Malignant Stem Cells. CURRENT STEM CELL REPORTS 2016; 2:379-387. [PMID: 28503404 DOI: 10.1007/s40778-016-0068-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Wnt signaling plays important roles in stem cell self-renewal and differentiation in adults as well as in embryonic development. Mutations that activate canonical Wnt/β-catenin signaling also initiate and maintain several cancer states, including colorectal cancer and leukemia, and hence Wnt inhibitors are currently being explored as therapeutic options. In this review, we summarize previous studies and update recent findings on canonical Wnt signaling and its components, as well as their roles in somatic stem cell homeostasis and maintenance of cancer initiating cells.
Collapse
Affiliation(s)
- Dheeraj Bhavanasi
- Department of Medicine (Hematology-Oncology), University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Peter S Klein
- Department of Medicine (Hematology-Oncology), University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.,Cell and Molecular Biology Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
197
|
Chin AM, Tsai YH, Finkbeiner SR, Nagy MS, Walker EM, Ethen NJ, Williams BO, Battle MA, Spence JR. A Dynamic WNT/β-CATENIN Signaling Environment Leads to WNT-Independent and WNT-Dependent Proliferation of Embryonic Intestinal Progenitor Cells. Stem Cell Reports 2016; 7:826-839. [PMID: 27720905 PMCID: PMC5106483 DOI: 10.1016/j.stemcr.2016.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 09/07/2016] [Accepted: 09/08/2016] [Indexed: 02/08/2023] Open
Abstract
Much of our understanding about how intestinal stem and progenitor cells are regulated comes from studying the late fetal stages of development and the adult intestine. In this light, little is known about intestine development prior to the formation of stereotypical villus structures with columnar epithelium, a stage when the epithelium is pseudostratified and appears to be a relatively uniform population of progenitor cells with high proliferative capacity. Here, we investigated a role for WNT/β-CATENIN signaling during the pseudostratified stages of development (E13.5, E14.5) and following villus formation (E15.5) in mice. In contrast to the well-described role for WNT/β-CATENIN signaling as a regulator of stem/progenitor cells in the late fetal and adult gut, conditional epithelial deletion of β-catenin or the Frizzled co-receptors Lrp5 and Lrp6 had no effect on epithelial progenitor cell proliferation in the pseudostratified epithelium. Mutant embryos displayed obvious developmental defects, including loss of proliferation and disruptions in villus formation starting only at E15.5. Mechanistically, our data suggest that WNT signaling-mediated proliferation at the time of villus formation is driven by mesenchymal, but not epithelial, WNT ligand secretion. WNT/β-CATENIN signaling is not required for proliferation during pseudostratified growth Deleting epithelial β-catenin causes loss of proliferation during villus morphogenesis Loss of WNT/β-CATENIN signaling leads to perturbations in villus formation Mesenchymal, not epithelial, WNT ligands are required for epithelial proliferation
Collapse
Affiliation(s)
- Alana M Chin
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yu-Hwai Tsai
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stacy R Finkbeiner
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Melinda S Nagy
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Emily M Walker
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Nicole J Ethen
- Program in Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Bart O Williams
- Program in Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Michele A Battle
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jason R Spence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
198
|
Lim Y, Kim CH, Lee SY, Kim H, Ahn SH, Lee SH, Koh JM, Rhee Y, Baek KH, Min YK, Kim DY, Kim BJ, Kang MI. Decreased Plasma Levels of Sclerostin But Not Dickkopf-1 are Associated with an Increased Prevalence of Osteoporotic Fracture and Lower Bone Mineral Density in Postmenopausal Korean Women. Calcif Tissue Int 2016; 99:350-9. [PMID: 27289555 DOI: 10.1007/s00223-016-0160-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/03/2016] [Indexed: 12/17/2022]
Abstract
Although sclerostin (SOST) and Dickkopf-related protein 1 (DKK1) are major regulators in bone metabolism, their associations with osteoporotic fracture (OF) in Asians are inconclusive. Furthermore, there have been no clinical studies separately considering non-vertebral and vertebral fractures in terms of the blood levels of SOST and DKK1. Among 513 consecutive postmenopausal Korean women, we identified 103 cases defined as subjects with OF (i.e., non-vertebral and/or vertebral fractures). The controls were randomly selected from the remaining 410 subjects and matched 1:1 to cases according to both age and body mass index. Non-vertebral and morphological vertebral fractures were identified by an interviewer-assisted questionnaire and lateral thoracolumbar radiographs, respectively. Bone mineral density (BMD) and plasma levels of SOST and DKK1 were measured. Plasma SOST levels were lower in subjects with OF than in the control group. Each standard deviation decrement of plasma SOST concentration was associated with a multivariable-adjusted odds ratio of 1.77 for any prevalent OF type. The odds for OF was 2.97-fold higher in subjects in the lowest SOST tertile compared with subjects in the highest SOST tertile. These associations remained significant when the non-vertebral and vertebral fractures were analyzed separately. However, prevalent OF was not associated with plasma DKK1 levels, regardless of the type of fracture and the adjustment model employed. Consistently, plasma SOST levels were positively related with BMD values at all measured skeletal sites, although this was not observed for DKK1. Circulating SOST but not DKK1 may be a potential biomarker for predicting bone health in Asians.
Collapse
Affiliation(s)
- Yejee Lim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, 222 Banpo-daero, Seocho-gu, Seoul, 137-701, Korea
| | - Chong Hwa Kim
- Department of Internal Medicine, Sejong General Hospital, Bucheon, 422-711, Korea
| | - Sun-Young Lee
- Asan Institute for Life Sciences, Seoul, 138-736, Korea
| | - Hyeonmok Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43 Gil, Songpa-Gu, Seoul, 138-736, Korea
| | - Seong Hee Ahn
- Department of Endocrinology, Inha University School of Medicine, Incheon, 22332, South Korea
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43 Gil, Songpa-Gu, Seoul, 138-736, Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43 Gil, Songpa-Gu, Seoul, 138-736, Korea
| | - Yumie Rhee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 120-752, Korea
| | - Ki Hyun Baek
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, 222 Banpo-daero, Seocho-gu, Seoul, 137-701, Korea
| | - Yong-Ki Min
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Deog-Yoon Kim
- Department of Nuclear Medicine, Kyunghee University School of Medicine, Seoul, 130-872, Korea
| | - Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43 Gil, Songpa-Gu, Seoul, 138-736, Korea.
| | - Moo-Il Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, 222 Banpo-daero, Seocho-gu, Seoul, 137-701, Korea.
| |
Collapse
|
199
|
Zhang X, Chen Y, Ye Y, Wang J, Wang H, Yuan G, Lin Z, Wu Y, Zhang Y, Lin X. Wnt signaling promotes hindgut fate commitment through regulating multi-lineage genes during hESC differentiation. Cell Signal 2016; 29:12-22. [PMID: 27693749 DOI: 10.1016/j.cellsig.2016.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 09/22/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022]
Abstract
Wnt signaling plays essential roles in both embryonic pattern formation and postembryonic tissue homoestasis. High levels of Wnt activity repress foregut identity and facilitate hindgut fate through forming a gradient of Wnt signaling activity along the anterior-posterior axis. Here, we examined the mechanisms of Wnt signaling in hindgut development by differentiating human embryonic stem cells (hESCs) into the hindgut progenitors. We observed severe morphological changes when Wnt signaling was blocked by using Wnt antagonist Dkk1. We performed deep-transcriptome sequencing (RNA-seq) and identified 240 Wnt-activated genes and 2023 Wnt-repressed genes, respectively. Clusters of Wnt targets showed enrichment in specific biological functions, such as "gastrointestinal or skeletal development" in the Wnt-activated targets and "neural or immune system development" in the Wnt-repressed targets. Moreover, we adopted a high-throughput chromatin immunoprecipitation and deep sequencing (ChIP-seq) approach to identify the genomic regions through which Wnt-activated transcription factor TCF7L2 regulated transcription. We identified 83 Wnt direct target candidates, including the hindgut marker CDX2 and the genes relevant to morphogenesis (MSX1, MSX2, LEF1, T, PDGFRB etc.) through combinatorial analysis of the RNA-seq and ChIP-seq data. Together, our study identified a series of direct and indirect Wnt targets in hindgut differentiation, and uncovered the diverse mechanisms of Wnt signaling in regulating multi-lineage differentiation.
Collapse
Affiliation(s)
- Xiujuan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ying Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ying Ye
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jianfeng Wang
- Core Genomic Facility, CAS Key Laboratory of Genome Sciences & Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Hong Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Guohong Yuan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhe Lin
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yihui Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xinhua Lin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Division of Developmental Biology, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States; State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
200
|
Zebisch M, Jackson VA, Zhao Y, Jones EY. Structure of the Dual-Mode Wnt Regulator Kremen1 and Insight into Ternary Complex Formation with LRP6 and Dickkopf. Structure 2016; 24:1599-605. [PMID: 27524201 PMCID: PMC5014086 DOI: 10.1016/j.str.2016.06.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 06/12/2016] [Accepted: 06/13/2016] [Indexed: 01/17/2023]
Abstract
Kremen 1 and 2 have been identified as co-receptors for Dickkopf (Dkk) proteins, hallmark secreted antagonists of canonical Wnt signaling. We present here three crystal structures of the ectodomain of human Kremen1 (KRM1ECD) at resolutions between 1.9 and 3.2 Å. KRM1ECD emerges as a rigid molecule with tight interactions stabilizing a triangular arrangement of its Kringle, WSC, and CUB structural domains. The structures reveal an unpredicted homology of the WSC domain to hepatocyte growth factor. We further report the general architecture of the ternary complex formed by the Wnt co-receptor Lrp5/6, Dkk, and Krm, determined from a low-resolution complex crystal structure between β-propeller/EGF repeats (PE) 3 and 4 of the Wnt co-receptor LRP6 (LRP6PE3PE4), the cysteine-rich domain 2 (CRD2) of DKK1, and KRM1ECD. DKK1CRD2 is sandwiched between LRP6PE3 and KRM1Kringle-WSC. Modeling studies supported by surface plasmon resonance suggest a direct interaction site between Krm1CUB and Lrp6PE2.
Collapse
Affiliation(s)
- Matthias Zebisch
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.
| | - Verity A Jackson
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Yuguang Zhao
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.
| |
Collapse
|