1001
|
Abstract
The Wnt pathway is a major embryonic signaling pathway that controls cell proliferation, cell fate, and body-axis determination in vertebrate embryos. Soon after egg fertilization, Wnt pathway components play a role in microtubule-dependent dorsoventral axis specification. Later in embryogenesis, another conserved function of the pathway is to specify the anteroposterior axis. The dual role of Wnt signaling in Xenopus and zebrafish embryos is regulated at different developmental stages by distinct sets of Wnt target genes. This review highlights recent progress in the discrimination of different signaling branches and the identification of specific pathway targets during vertebrate axial development.
Collapse
Affiliation(s)
- Hiroki Hikasa
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | | |
Collapse
|
1002
|
Estrogen regulation of Dkk1 and Wnt/β-Catenin signaling in neurodegenerative disease. Brain Res 2012; 1514:63-74. [PMID: 23261660 DOI: 10.1016/j.brainres.2012.12.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/11/2012] [Indexed: 11/22/2022]
Abstract
17β-estradiol (E2 or estrogen) is an endogenous steroid hormone that is well known to exert neuroprotection. Along these lines, one mechanism through which E2 protects the hippocampus from cerebral ischemia is by preventing the post-ischemic elevation of Dkk1, a neurodegenerative factor that serves as an antagonist of the canonical Wnt signaling pathway, and simultaneously inducing pro-survival Wnt/β-Catenin signaling in hippocampal neurons. Intriguingly, while expression of Dkk1 is required for proper neural development, overexpression of Dkk1 is characteristic of many neurodegenerative diseases, such as stroke, Alzheimer's disease, Parkinson's disease, and temporal lobe epilepsy. In this review, we will briefly summarize the canonical Wnt signaling pathway, highlight the current literature linking alterations of Dkk1 and Wnt/β-Catenin signaling with neurological disease, and discuss E2's role in maintaining the delicate balance of Dkk1 and Wnt/β-Catenin signaling in the adult brain. Finally, we will consider the implications of long-term E2 deprivation and hormone therapy on this crucial neural pathway. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
|
1003
|
Kumawat K, Menzen MH, Bos IST, Baarsma HA, Borger P, Roth M, Tamm M, Halayko AJ, Simoons M, Prins A, Postma DS, Schmidt M, Gosens R. Noncanonical WNT-5A signaling regulates TGF-β-induced extracellular matrix production by airway smooth muscle cells. FASEB J 2012; 27:1631-43. [PMID: 23254341 DOI: 10.1096/fj.12-217539] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Transforming growth factor β (TGF-β), a key mediator of fibrotic responses, is increased in asthma and drives airway remodeling by inducing expression of extracellular matrix (ECM) proteins. We investigated the molecular mechanisms underlying TGF-β-induced ECM expression by airway smooth muscle cells and demonstrate a novel link between TGF-β and Wingless/integrase 1 (WNT) signaling in ECM deposition. Airway smooth muscle expresses abundant WNT ligands, with the noncanonical WNT-5A being the most profoundly expressed. Interestingly, WNT-5A shows ∼2-fold higher abundance in airway smooth muscle cells isolated from individuals with asthma than individuals without asthma. WNT-5A is markedly induced in response to TGF-β (4-16-fold; EC₅₀ 0.3 ng/ml) and is required for collagen and fibronectin expression by airway smooth muscle. WNT-5A engages noncanonical WNT signaling pathways, as inhibition of Ca(2+) and c-Jun N-terminal kinase (JNK) signaling attenuated this TGF-β response, whereas the canonical WNT antagonist Dickkopf 1 (DKK-1) did not. Accordingly, WNT-5A induced JNK phosphorylation and nuclear translocation of nuclear factor of activated T cells c1 (NFATc1). Furthermore, silencing of the WNT-5A receptors Frizzled 8 (FZD8) and RYK attenuated TGF-β-induced ECM expression. Collectively, these findings demonstrate that noncanonical WNT-5A signaling is activated by and necessary for TGF-β-induced ECM production by airway smooth muscle cells, which could have significance in asthma pathogenesis.
Collapse
Affiliation(s)
- Kuldeep Kumawat
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1004
|
Wnt Signaling in Neurogenesis during Aging and Physical Activity. Brain Sci 2012; 2:745-68. [PMID: 24961268 PMCID: PMC4061808 DOI: 10.3390/brainsci2040745] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 11/27/2012] [Accepted: 12/10/2012] [Indexed: 12/22/2022] Open
Abstract
Over the past decade, much progress has been made regarding our understanding of neurogenesis in both young and old animals and where it occurs throughout the lifespan, although the growth of new neurons declines with increasing age. In addition, physical activity can reverse this age-dependent decline in neurogenesis. Highly correlated with this decline is the degree of inter and intracellular Wnt signaling, the molecular mechanisms of which have only recently started to be elucidated. So far, most of what we know about intracellular signaling during/following exercise centers around the CREB/CRE initiated transcriptional events. Relatively little is known, however, about how aging and physical activity affect the Wnt signaling pathway. Herein, we briefly review the salient features of neurogenesis in young and then in old adult animals. Then, we discuss Wnt signaling and review the very few in vitro and in vivo studies that have examined the Wnt signaling pathways in aging and physical activity.
Collapse
|
1005
|
Fukuma M, Tanese K, Effendi K, Yamazaki K, Masugi Y, Suda M, Sakamoto M. Leucine-rich repeat-containing G protein-coupled receptor 5 regulates epithelial cell phenotype and survival of hepatocellular carcinoma cells. Exp Cell Res 2012; 319:113-21. [PMID: 23127514 DOI: 10.1016/j.yexcr.2012.10.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 10/11/2012] [Accepted: 10/26/2012] [Indexed: 11/19/2022]
Abstract
The leucine-rich repeat containing G protein-coupled receptor 5 (LGR5), also known as GPR49, is a seven-transmembrane receptor that is expressed in stem cells of the intestinal crypts and hair follicles of mice. LGR5 is overexpressed in some types of human cancer, and is one of the target genes of the Wnt signaling pathway. To explore the function of LGR5 in cancer cells, stable hepatocellular carcinoma (HCC) cell lines expressing FLAG-tagged LGR5 were established. Overexpression of LGR5 resulted in changes in cell shape from an extended flat (mesenchymal) phenotype to a round aggregated (stem cell-like) phenotype. Cells transfected with LGR5 showed higher colony forming activity, and were more resistant to a cytotoxic drug than cells transfected with empty vector. Overexpression of LGR5 inhibited cell motility. LGR5-transfected cells formed nodule type tumors in the livers of immunodeficient mice, whereas empty vector-transfected cells formed more invasive tumors. Down-regulation of LGR5 changed the morphology of HCC cells from the aggregated phenotype to an extended spindle phenotype, and cell motility was increased. This is the first study reporting the functional role of LGR5 in the biology of HCC cells, and the results suggest that aberrant expression of LGR5 regulates epithelial cell phenotype and survival.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Cell Survival/genetics
- Cells, Cultured
- Epithelial Cells/metabolism
- Epithelial Cells/physiology
- Gene Expression Regulation, Neoplastic
- Hep G2 Cells
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Phenotype
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/physiology
Collapse
Affiliation(s)
- Mariko Fukuma
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
1006
|
Saran U, Arfuso F, Zeps N, Dharmarajan A. Secreted frizzled-related protein 4 expression is positively associated with responsiveness to cisplatin of ovarian cancer cell lines in vitro and with lower tumour grade in mucinous ovarian cancers. BMC Cell Biol 2012; 13:25. [PMID: 23039795 PMCID: PMC3521476 DOI: 10.1186/1471-2121-13-25] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 10/01/2012] [Indexed: 12/29/2022] Open
Abstract
Background Ovarian cancer is one of the most lethal malignancies in women, as it is frequently detected at an advanced stage, and cancers often become refractory to chemotherapy. Evidence suggests that dysregulation of pro-apoptotic genes plays a key role in the onset of chemoresistance. The secreted Frizzled-Related Protein (sFRP) family is pro-apoptotic and also a negative modulator of the Wnt signalling cascade. Studies have demonstrated that the re-expression of sFRPs, in particular sFRP4, is associated with a better prognosis, and that experimentally induced expression results in cell death. Results In vitro experimental models determined that sFRP4 was differentially expressed in chemosensitive (A2780) and chemoresistant (A2780 ADR and A2780 Cis) ovarian cell lines, with chemosensitive cells expressing significantly higher levels of sFRP4. Transfection of the chemoresistant cell lines with sFRP4 significantly increased their sensitivity to chemotherapy. Conversely, silencing of sFRP4 expression in the chemosensitive cell line resulted in a corresponding increase in chemoresistance. Comparison of sFRP4 expression in tumour biopsies revealed a positive trend between sFRP4 expression and tumour grade, with mucinous cyst adenocarcinomas exhibiting significantly decreased sFRP4 levels compared to mucinous borderline tumours. Conclusions This study indicates a role for sFRP4 as a predictive marker of chemosensitivity in ovarian cancer and suggests that this pathway may be worth exploiting for novel therapies.
Collapse
Affiliation(s)
- Uttara Saran
- School of Anatomy and Human Biology, Faculty of Life and Physical Sciences, The University of Western Australia, Perth, Crawley, Western Australia
| | | | | | | |
Collapse
|
1007
|
Olguín HC, Pisconti A. Marking the tempo for myogenesis: Pax7 and the regulation of muscle stem cell fate decisions. J Cell Mol Med 2012; 16:1013-25. [PMID: 21615681 PMCID: PMC4365881 DOI: 10.1111/j.1582-4934.2011.01348.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Post-natal growth and regeneration of skeletal muscle is highly dependent on a population of resident myogenic precursors known as satellite cells. Transcription factors from the Pax gene family, Pax3 and Pax7, are critical for satellite cell biogenesis, survival and potentially self-renewal; however, the underlying molecular mechanisms remain unsolved. This is particularly true in the case of Pax7, which appears to regulate myogenesis at multiple levels. Accordingly, recent data have highlighted the importance of a functional relationship between Pax7 and the MyoD family of muscle regulatory transcription factors during normal muscle formation and disease. Here we will critically review key findings suggesting that Pax7 may play a dual role by promoting resident muscle progenitors to commit to the skeletal muscle lineage while preventing terminal differentiation, thus keeping muscle progenitors poised to differentiate upon environmental cues. In addition, potential regulatory mechanisms for the control of Pax7 activity will be proposed.
Collapse
Affiliation(s)
- Hugo C Olguín
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | |
Collapse
|
1008
|
Zhu J, Zhang S, Gu L, Di W. Epigenetic silencing of DKK2 and Wnt signal pathway components in human ovarian carcinoma. Carcinogenesis 2012; 33:2334-43. [PMID: 22964660 DOI: 10.1093/carcin/bgs278] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Wnt/β-Catenin signaling dysregulation is involved in tumorigenesis. Furthermore, epigenetic modification of the Dickkopf (DKK) family (DKK1-DKK4) has been shown to be important in Wnt signaling regulation. In this study, the role of DKK2, a Wnt antagonist, in epithelial ovarian cancer (EOC) was evaluated by examining the expression and methylation of DKK2 in SKOV3 and ES-2 ovarian cancer cell lines and 78 tissues collected from patients (50 ovarian carcinoma, 20 benign tumor and 8 normal ovarian tissues). DKK2 is highly downregulated in EOCs; however, DKK2 expression levels are higher in both normal tissues and benign tumors. In most cases of ovarian carcinoma, DKK2 is methylated, compared with the more common unmethylated form present in benign tumors and normal ovarian tissues. Additionally, DKK2 may be epigenetically silenced by methylation in higher grades and stages of EOC. Functional analysis revealed that overexpression of DKK2 suppressed malignant cell growth and invasion in SKOV3 and ES-2 cell lines. The expression of the downstream genes of Wnt signaling, including β-catenin, c-Myc and cyclin D1, was decreased in DKK2-transfected cells compared with mock cells. The expression of matrix metalloproteinase-2 and focal adhesion kinase were also decreased in DKK2 transfectants, supporting findings indicating inhibition of cell migration and invasion. This report provides novel indications that DKK2 is a unique hypermethylated target gene in EOC and that DKK2 may contribute to tumorigenesis in EOC through the Wnt/β-catenin signaling mechanisms.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Obstetrics and Gynecology, Ren-Ji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200127, China
| | | | | | | |
Collapse
|
1009
|
Sani G, Napoletano F, Forte AM, Kotzalidis GD, Panaccione I, Porfiri GM, Simonetti A, Caloro M, Girardi N, Telesforo CL, Serra G, Romano S, Manfredi G, Savoja V, Tamorri SM, Koukopoulos AE, Serata D, Rapinesi C, Casale AD, Nicoletti F, Girardi P. The wnt pathway in mood disorders. Curr Neuropharmacol 2012; 10:239-253. [PMID: 23449817 PMCID: PMC3468878 DOI: 10.2174/157015912803217279] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 03/13/2012] [Accepted: 03/24/2012] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES To review the evidence of the involvement of the Wnt signalling pathway in mood disorders and in the action of drugs used to treat these disorders. METHODS We performed a careful PubMed search using as keywords all possible terms relevant to the Wnt pathway and crossing them with each of four areas, i.e., developmental effects, behavioural effects, mood disorders, and drugs used in their treatment. Papers were selected on the basis of their content and their data used for discussion. RESULTS Neurodevelopmental and behavioural data point to the possibility of involvement of the Wnt pathway in the pathophysiology of mood disorders. Clinical and post-mortem data are not sufficient to corroborate a definite role for Wnt alterations in any mood disorder. Combining genetic and pharmacological data, we may state that glycogen synthase kinase is the key molecule in bipolar disorder, as it is connected with many other signalling pathways that were shown to be involved in mood disorders, while Wnt molecules in the hippocampus appear to be mainly involved in depressive disorders. CONCLUSIONS Altered Wnt signalling may play a role in the pathophysiology of mood disorders, although not a central one. It is premature to draw conclusions regarding the possible usefulness of Wnt manipulations in the treatment of mood disorders.
Collapse
Affiliation(s)
- Gabriele Sani
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
- Centro Lucio Bini, Rome, Italy
| | - Flavia Napoletano
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Alberto Maria Forte
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
- NEUROMED, Pozzilli, Isernia, Italy
| | - Giorgio D Kotzalidis
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Isabella Panaccione
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
- NEUROMED, Pozzilli, Isernia, Italy
| | - Giulio Maria Porfiri
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Alessio Simonetti
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
- Centro Lucio Bini, Rome, Italy
| | - Matteo Caloro
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Nicoletta Girardi
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Carla Ludovica Telesforo
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Giulia Serra
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
- Centro Lucio Bini, Rome, Italy
| | - Silvia Romano
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Giovanni Manfredi
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Valeria Savoja
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Stefano Maria Tamorri
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Alexia E Koukopoulos
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
- Centro Lucio Bini, Rome, Italy
| | - Daniele Serata
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Chiara Rapinesi
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
- Department of Neuropsychiatry, Villa Rosa, Suore Hospitaliere of the Sacred Heart of Jesus, Viterbo, Italy
| | - Antonio Del Casale
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Ferdinando Nicoletti
- NEUROMED, Pozzilli, Isernia, Italy
- Department of Neuropharmacology, Sapienza University, School of Medicine and Pharmacy, Rome, Italy
| | - Paolo Girardi
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| |
Collapse
|
1010
|
Vanuytsel T, Senger S, Fasano A, Shea-Donohue T. Major signaling pathways in intestinal stem cells. Biochim Biophys Acta Gen Subj 2012; 1830:2410-26. [PMID: 22922290 DOI: 10.1016/j.bbagen.2012.08.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 07/05/2012] [Accepted: 08/07/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND The discovery of markers to identify the intestinal stem cell population and the generation of powerful transgenic mouse models to study stem cell physiology have led to seminal discoveries in stem cell biology. SCOPE OF REVIEW In this review we give an overview of the current knowledge in the field of intestinal stem cells (ISCs) highlighting the most recent progress on markers defining the ISC population and pathways governing intestinal stem cell maintenance and differentiation. Furthermore we review their interaction with other stem cell related pathways. Finally we give an overview of alteration of these pathways in human inflammatory gastrointestinal diseases. MAJOR CONCLUSIONS We highlight the complex network of interactions occurring among different pathways and put in perspective the many layers of regulation that occur in maintaining the intestinal homeostasis. GENERAL SIGNIFICANCE Understanding the involvement of ISCs in inflammatory diseases can potentially lead to new therapeutic approaches to treat inflammatory GI pathologies such as IBD and celiac disease and could reveal the molecular mechanisms leading to the pathogenesis of dysplasia and cancer in inflammatory chronic conditions. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Tim Vanuytsel
- Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
1011
|
Chang H, Balenci L, Okolowsky N, Muller WJ, Hamel PA. Mammary epithelial-restricted expression of activated c-src rescues the block to mammary gland morphogenesis due to the deletion of the C-terminus of Patched-1. Dev Biol 2012; 370:187-97. [PMID: 22968113 DOI: 10.1016/j.ydbio.2012.07.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 07/25/2012] [Accepted: 07/26/2012] [Indexed: 12/26/2022]
Abstract
Mesenchymal dysplasia (mes) mice expressing a C-terminally truncated version of the Hedgehog (Hh)-ligand receptor, Patched-1 (Ptch1), exhibit a limited spectrum of developmental defects including blocked ductal morphogenesis of the mammary gland during puberty. Given that the Hh-ligands can stimulate signalling cascades distinct from the canonical pathway involving Smo and the Gli-family proteins and that Ptch1 binds to factors harbouring SH3-domains, we determined whether the mes mammary gland defect could be rescued by activating non-canonical signalling pathways downstream of Ptch1. We demonstrate here that expression of constitutively active c-src (c-src(Act)) in mammary epithelial cells overcomes the block to mammary epithelial morphogenesis in mes mice. Specifically, MMTV-directed expression of c-src(Act) rescued blocked ductal morphogenesis in mes mice, albeit only after animals were more than 15 weeks of age. The overall morphology resembled wild type mice expressing c-src(Act) although 40% of mes/MMTV-c-src(Act) mice exhibited terminal end buds at 24 weeks of age. C-src(Act) restored the proliferative capacity of mes epithelial cells, self-renewal capacity of mammary progenitor cells and increased the expression of Esr1, Ptch1 and Gli1. These data reveal the cooperative interactions between signalling cascades involving c-src and Ptch1 and suggest that Hh-signalling may be permissive for c-src/Esr1-dependent mammary gland morphogenesis.
Collapse
Affiliation(s)
- Hong Chang
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Canada
| | | | | | | | | |
Collapse
|
1012
|
Roy S, Majumdar AP. Signaling in colon cancer stem cells. J Mol Signal 2012; 7:11. [PMID: 22866952 PMCID: PMC3485105 DOI: 10.1186/1750-2187-7-11] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/11/2012] [Indexed: 02/07/2023] Open
Abstract
: Colorectal cancer is the fourth most common form of cancer worldwide and ranks third among the cancer-related deaths in the US and other Western countries. It occurs with equal frequency in men and women, constituting 10% of new cancer cases in men and 11% in women. Despite recent advancement in therapeutics, the survival rates from metastatic are less than 5%. Growing evidence supports the contention that epithelial cancers including colorectal cancer, the incidence of which increases with aging, are diseases driven by the pluripotent, self-renewing cancer stem cells (CSCs). Dysregulation of Wnt, Notch, Hedgehog and/or TGF-β signaling pathways that are involved in proliferation and maintenance of CSCs leads to the development of CRC. This review focuses on the signaling pathways relevant for CRC to understand the mechanisms leading to tumor progression and therapy resistance, which may help in the development of therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Sanchita Roy
- John D Dingell VA Medical Centre, 4646 John R; Room: B-4238, Detroit, MI, 48201, USA.
| | | |
Collapse
|
1013
|
Cooper SJ, von Roemeling CA, Kang KH, Marlow LA, Grebe SK, Menefee ME, Tun HW, Colon-Otero G, Perez EA, Copland JA. Reexpression of tumor suppressor, sFRP1, leads to antitumor synergy of combined HDAC and methyltransferase inhibitors in chemoresistant cancers. Mol Cancer Ther 2012; 11:2105-15. [PMID: 22826467 DOI: 10.1158/1535-7163.mct-11-0873] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Metastatic solid tumors are aggressive and mostly drug resistant, leading to few treatment options and poor prognosis as seen with clear cell renal cell carcinoma (ccRCC) and triple-negative breast cancer (TNBC). Therefore, the identification of new therapeutic regimes for the treatment of metastatic disease is desirable. ccRCC and TNBC cell lines were treated with the HDAC inhibitor romidepsin and the methyltransferase inhibitor decitabine, two epigenetic modifying drugs approved by the U.S. Food and Drug Administration for the treatment of various hematologic malignancies. Cell proliferation analysis, flow cytometry, quantitative PCR, and immunoblotting techniques were used to evaluate the antitumor synergy of this drug combination and identify the reexpression of epigenetically silenced tumor suppressor genes. Combinatorial treatment of metastatic TNBC and stage IV ccRCC cell lines with romidepsin/decitabine leads to synergistic inhibition of cell growth and induction of apoptosis above levels of individual drug treatments alone. Synergistic reexpression of the tumor suppressor gene secreted frizzled-related protein one (sFRP1) was observed in combinatorial drug-treated groups. Silencing sFRP1 (short hairpin RNA) before combinatorial drug treatment showed that sFRP1 mediates the growth inhibitory and apoptotic activity of combined romidepsin/decitabine. Furthermore, addition of recombinant sFRP1 to ccRCC or TNBC cells inhibits cell growth in a dose-dependent manner through the induction of apoptosis, identifying that epigenetic silencing of sFRP1 contributes to renal and breast cancer cell survival. Combinatorial treatment with romidepsin and decitabine in drug resistant tumors is a promising treatment strategy. Moreover, recombinant sFRP1 may be a novel therapeutic strategy for cancers with suppressed sFRP1 expression.
Collapse
Affiliation(s)
- Simon J Cooper
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1014
|
Bolzoni M, Donofrio G, Storti P, Guasco D, Toscani D, Lazzaretti M, Bonomini S, Agnelli L, Capocefalo A, Dalla Palma B, Neri A, Nicolini F, Lisignoli G, Russo F, Colla S, Aversa F, Giuliani N. Myeloma cells inhibit non-canonical wnt co-receptor ror2 expression in human bone marrow osteoprogenitor cells: effect of wnt5a/ror2 pathway activation on the osteogenic differentiation impairment induced by myeloma cells. Leukemia 2012; 27:451-63. [PMID: 22781592 DOI: 10.1038/leu.2012.190] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multiple myeloma (MM) is characterized by the impaired osteogenic differentiation of human mesenchymal stromal cells (hMSCs). Canonical Wnt signaling is critical for the regulation of bone formation, however, recent evidence suggests that the non-canonical Wnt agonist Wnt5a stimulates human osteoblastogenesis through its co-receptor Ror2. The effects of MM cells on non-canonical Wnt signaling and the effect of the activation of this pathway on MM-induced osteoblast exhaustion are not known and were investigated in this study. We found that the osteogenic differentiation of bone marrow hMSCs toward osteoprogenitor cells (PreOB) significantly increased Ror2 expression, and that MM cells inhibit Ror2 expression by PreOB in co-culture by inhibiting the non-canonical Wnt5a signaling. The activation of the non-canonical Wnt pathway in hMSCs by means of Wnt5a treatment and the overexpression of Wnt5 or Ror2 by lentiviral vectors increased the osteogenic differentiation of hMSCs and blunted the inhibitory effect of MM in co-culture. Consistently, Wnt5a inhibition by specific small interfering RNA reduced the hMSC expression of osteogenic markers. Our findings demonstrate that the Wnt5a/Ror2 pathway is involved in the pathophysiology of MM-induced bone disease and that the activation of the non-canonical Wnt5a/Ror2 pathway in hMSCs increases osteogenic differentiation and may counterbalance the inhibitory effect of MM cells.
Collapse
Affiliation(s)
- M Bolzoni
- Hematology and BMT Center, University of Parma, Parma, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1015
|
van Amerongen R, Fuerer C, Mizutani M, Nusse R. Wnt5a can both activate and repress Wnt/β-catenin signaling during mouse embryonic development. Dev Biol 2012; 369:101-14. [PMID: 22771246 PMCID: PMC3435145 DOI: 10.1016/j.ydbio.2012.06.020] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 06/20/2012] [Accepted: 06/27/2012] [Indexed: 01/04/2023]
Abstract
Embryonic development is controlled by a small set of signal transduction pathways, with vastly different phenotypic outcomes depending on the time and place of their recruitment. How the same molecular machinery can elicit such specific and distinct responses, remains one of the outstanding questions in developmental biology. Part of the answer may lie in the high inherent genetic complexity of these signaling cascades, as observed for the Wnt-pathway. The mammalian genome encodes multiple Wnt proteins and receptors, each of which show dynamic and tightly controlled expression patterns in the embryo. Yet how these components interact in the context of the whole organism remains unknown. Here we report the generation of a novel, inducible transgenic mouse model that allows spatiotemporal control over the expression of Wnt5a, a protein implicated in many developmental processes and multiple Wnt-signaling responses. We show that ectopic Wnt5a expression from E10.5 onwards results in a variety of developmental defects, including loss of hair follicles and reduced bone formation in the skull. Moreover, we find that Wnt5a can have dual signaling activities during mouse embryonic development. Specifically, Wnt5a is capable of both inducing and repressing β-catenin/TCF signaling in vivo, depending on the time and site of expression and the receptors expressed by receiving cells. These experiments show for the first time that a single mammalian Wnt protein can have multiple signaling activities in vivo, thereby furthering our understanding of how signaling specificity is achieved in a complex developmental context.
Collapse
Affiliation(s)
- Renée van Amerongen
- Department of Developmental Biology and Howard Hughes Medical Institute, Lorry I. Lokey Stem Cell Research Building, Stanford University, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
1016
|
Castañon BI, Stapp AD, Gifford CA, Spicer LJ, Hallford DM, Hernandez Gifford JA. Follicle-stimulating hormone regulation of estradiol production: possible involvement of WNT2 and β-catenin in bovine granulosa cells. J Anim Sci 2012; 90:3789-97. [PMID: 22696613 DOI: 10.2527/jas.2011-4696] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Follicle-stimulating hormone regulation of estrogen biosynthesis in the adult rodent ovary requires β-catenin (CTNNB1), but whether CTNNB1 is involved in FSH-induced estrogen production in cattle is unknown. To elucidate the effect of FSH in regulating specific wingless-type mouse mammary tumor virus integration site (WNT)/CTNNB1 pathway components in bovine folliculogenesis and steroidogenesis, granulosa cells and follicular fluid were collected from large antral follicles (8 to 22 mm) from ovaries containing stage-III corpora lutea (d 11 to 17 of an estrous cycle). Follicles were categorized as high estradiol (n = 3; ≥ 25 ng/mL) or low estradiol (n = 3; ≤ 14 ng/mL) based on intra-follicular estradiol concentrations. Protein fractions were collected from granulosa cells and CTNNB1 abundance was analyzed by Western blot. Follicles with increased estradiol concentrations had 6-fold greater (P < 0.001) abundances of CTNNB1 compared with those classified as low-estradiol follicles, indicating that the hormonal milieu responsible for increased estradiol content could result in CTNNB1 accumulation. To ascertain specific contributions of FSH to increases in CTNNB1 protein abundances, granulosa cells were isolated from small ovarian follicles (1 to 5 mm) and cultured in the presence or absence of 100 ng/mL FSH for 24 or 48 h. Real-time PCR quantification of aromatase (CYP19A1) and select WNT family members were evaluated in response to FSH treatment. Successful stimulation of granulosa cells with FSH was confirmed by induction of CYP19A1 mRNA and parallel temporal increases of medium estradiol concentrations. Additionally, protein kinase b (AKT), a known FSH target, increased 1.7-fold (P = 0.07). Of the WNT family members analyzed, only WNT2 mRNA was induced after 24 h of FSH treatment compared with controls (0.12-fold and 3.7-fold for control and FSH-treated, respectively; P < 0.05), and WNT2 expression tended (P = 0.11) to remain increased at 48 h in FSH-treated cells compared with controls (1.0- and 3.14-fold, respectively). Furthermore, FSH-treated granulosa cells had greater abundances of total CTNNB1 (P = 0.04) protein. These data demonstrate for the first time that FSH regulates CTNNB1 protein and WNT2 mRNA expressions in bovine granulosa cells, suggesting a potential role of canonical WNT signaling in ovarian steroidogenesis and follicular growth of cattle. Future studies are necessary to determine if FSH directly regulates CTNNB1 through modulation of AKT or indirectly by up regulating WNT2, which subsequently activates the canonical WNT pathway.
Collapse
Affiliation(s)
- B I Castañon
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA
| | | | | | | | | | | |
Collapse
|
1017
|
Abstract
B2A (B2A2-K-NS) is a synthetic multi-domain peptide that in vitro augments bone morphogenetic protein (BMP)-2-induced cell responsiveness and osteodifferentiation. Augmentation of endogenous BMP-2 is thought to ultimately improve bone repair, and has led to clinical evaluation of B2A in orthopedic applications. In this study, we show that B2A binds to BMP receptor (BMPR)-IB, BMPR-II, and BMPR-IA. B2A reduces the EC50 of rh-BMP-2, thus shifting the response curve to the left. B2A enhances the osteogenic activity of BMP-2, but not growth and differentiation factor-5, BMP-7, or BMP-9, indicating its action is highly BMP-2 selective. Additionally, B2A did not augment Wnt-3a- and retinoic acid-induced differentiation. All three functional domains (receptor-binding domain, hydrophobic-linker domain, heparin-binding domain) of B2A are required for optimal bioactivity. Collectively, the results suggest that B2A, via its unique sequence, acts in a manner consistent with a positive receptor modulator to selectively enhance BMP-2 osteodifferentiation, and yet in the absence of BMP-2, B2A is without cooperative effect.
Collapse
Affiliation(s)
- Xinhua Lin
- BioSurface Engineering Technologies, Inc., Rockville, MD 20850, USA.
| | | | | | | | | |
Collapse
|
1018
|
Yeung CH, Wang K, Cooper TG. Why are epididymal tumours so rare? Asian J Androl 2012; 14:465-75. [PMID: 22522502 DOI: 10.1038/aja.2012.20] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Epididymal tumour incidence is at most 0.03% of all male cancers. It is an enigma why the human epididymis does not often succumb to cancer, when it expresses markers of stem and cancer cells, and constitutively expresses oncogenes, pro-proliferative and pro-angiogenic factors that allow tumour cells to escape immunosurveillance in cancer-prone tissues. The privileged position of the human epididymis in evading tumourigenicity is reflected in transgenic mouse models in which induction of tumours in other organs is not accompanied by epididymal neoplasia. The epididymis appears to: (i) prevent tumour initiation (it probably lacks stem cells and has strong anti-oxidative mechanisms, active tumour suppressors and inactive oncogene products); (ii) foster tumour monitoring and destruction (by strong immuno-surveillance and -eradication, and cellular senescence); (iii) avert proliferation and angiogenesis (with persistent tight junctions, the presence of anti-angiogenic factors and misplaced pro-angiogenic factors), which together (iv) promote dormancy and restrict dividing cells to hyperplasia. Epididymal cells may be rendered non-responsive to oncogenic stimuli by the constitutive expression of factors generally inducible in tumours, and resistant to the normal epididymal environment, which mimics that of a tumour niche promoting tumour growth. The threshold for tumour initiation may thus be higher in the epididymis than in other organs. Several anti-tumour mechanisms are those that maintain spermatozoa quiescent and immunologically silent, so the low incidence of cancer in the epididymis may be a consequence of its role in sperm maturation and storage. Understanding these mechanisms may throw light on cancer prevention and therapy in general.
Collapse
Affiliation(s)
- Ching-Hei Yeung
- Shandong Stem Cell Engineering and Technology Research Centre, YuHuangDing Hospital, Yantai, China
| | | | | |
Collapse
|
1019
|
Abstract
Excessive caloric intake without a rise in energy expenditure promotes adipocyte hyperplasia and adiposity. The rise in adipocyte number is triggered by signaling factors that induce conversion of mesenchymal stem cells (MSCs) to preadipocytes that differentiate into adipocytes. MSCs, which are recruited from the vascular stroma of adipose tissue, provide an unlimited supply of adipocyte precursors. Members of the BMP and Wnt families are key mediators of stem cell commitment to produce preadipocytes. Following commitment, exposure of growth-arrested preadipocytes to differentiation inducers [insulin-like growth factor 1 (IGF1), glucocorticoid, and cyclic AMP (cAMP)] triggers DNA replication and reentry into the cell cycle (mitotic clonal expansion). Mitotic clonal expansion involves a transcription factor cascade, followed by the expression of adipocyte genes. Critical to these events are phosphorylations of the transcription factor CCATT enhancer-binding protein β (C/EBPβ) by MAP kinase and GSK3β to produce a conformational change that gives rise to DNA-binding activity. "Activated" C/EBPβ then triggers transcription of peroxisome proliferator-activated receptor-γ (PPARγ) and C/EBPα, which in turn coordinately activate genes whose expression produces the adipocyte phenotype.
Collapse
Affiliation(s)
- Qi Qun Tang
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
1020
|
Sivakumar KC, Dhanesh SB, Shobana S, James J, Mundayoor S. A systems biology approach to model neural stem cell regulation by notch, shh, wnt, and EGF signaling pathways. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2012; 15:729-37. [PMID: 21978399 DOI: 10.1089/omi.2011.0011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The Notch, Sonic Hedgehog (Shh), Wnt, and EGF pathways have long been known to influence cell fate specification in the developing nervous system. Here we attempted to evaluate the contemporary knowledge about neural stem cell differentiation promoted by various drug-based regulations through a systems biology approach. Our model showed the phenomenon of DAPT-mediated antagonism of Enhancer of split [E(spl)] genes and enhancement of Shh target genes by a SAG agonist that were effectively demonstrated computationally and were consistent with experimental studies. However, in the case of model simulation of Wnt and EGF pathways, the model network did not supply any concurrent results with experimental data despite the fact that drugs were added at the appropriate positions. This paves insight into the potential of crosstalks between pathways considered in our study. Therefore, we manually developed a map of signaling crosstalk, which included the species connected by representatives from Notch, Shh, Wnt, and EGF pathways and highlighted the regulation of a single target gene, Hes-1, based on drug-induced simulations. These simulations provided results that matched with experimental studies. Therefore, these signaling crosstalk models complement as a tool toward the discovery of novel regulatory processes involved in neural stem cell maintenance, proliferation, and differentiation during mammalian central nervous system development. To our knowledge, this is the first report of a simple crosstalk map that highlights the differential regulation of neural stem cell differentiation and underscores the flow of positive and negative regulatory signals modulated by drugs.
Collapse
|
1021
|
Ossipova O, Sokol SY. Neural crest specification by noncanonical Wnt signaling and PAR-1. Development 2012; 138:5441-50. [PMID: 22110058 DOI: 10.1242/dev.067280] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neural crest (NC) cells are multipotent progenitors that form at the neural plate border, undergo epithelial-mesenchymal transition and migrate to diverse locations in vertebrate embryos to give rise to many cell types. Multiple signaling factors, including Wnt proteins, operate during early embryonic development to induce the NC cell fate. Whereas the requirement for the Wnt/β-catenin pathway in NC specification has been well established, a similar role for Wnt proteins that do not stabilize β-catenin has remained unclear. Our gain- and loss-of-function experiments implicate Wnt11-like proteins in NC specification in Xenopus embryos. In support of this conclusion, modulation of β-catenin-independent signaling through Dishevelled and Ror2 causes predictable changes in premigratory NC. Morpholino-mediated depletion experiments suggest that Wnt11R, a Wnt protein that is expressed in neuroectoderm adjacent to the NC territory, is required for NC formation. Wnt11-like signals might specify NC by altering the localization and activity of the serine/threonine polarity kinase PAR-1 (also known as microtubule-associated regulatory kinase or MARK), which itself plays an essential role in NC formation. Consistent with this model, PAR-1 RNA rescues NC markers in embryos in which noncanonical Wnt signaling has been blocked. These experiments identify novel roles for Wnt11R and PAR-1 in NC specification and reveal an unexpected connection between morphogenesis and cell fate.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
1022
|
Wilson NH, Stoeckli ET. Sonic Hedgehog regulates Wnt activity during neural circuit formation. VITAMINS AND HORMONES 2012; 88:173-209. [PMID: 22391304 DOI: 10.1016/b978-0-12-394622-5.00008-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gradients of secreted morphogens, such as Sonic hedgehog (Shh), Wnt, and TGFβ/Bmp, have classically been shown to control many aspects of early development by regulating cell proliferation and determining cell fate. However, recent studies demonstrate that these molecules also play important and evolutionarily conserved roles in later aspects of neural development. Depending on the context, these molecules can elicit gene transcription in the nucleus, or alternatively can provide instructive signals at the growth cone that induce local and rapid changes in cytoskeletal organization. Shh can activate different cellular transduction pathways via its binding to alternative coreceptor complexes or simply by adaptation of its "classical" signaling pathway. However, in most of its activities during neural development, Shh does not act alone but rather in concert with other morphogens, particularly the Wnts. This review provides an overview of the mechanisms by which Shh signaling acts in concert with Wnts to mediate a myriad of cellular processes that are required for neural circuit formation.
Collapse
Affiliation(s)
- Nicole H Wilson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
1023
|
Hasegawa K, Yasuda SY, Teo JL, Nguyen C, McMillan M, Hsieh CL, Suemori H, Nakatsuji N, Yamamoto M, Miyabayashi T, Lutzko C, Pera MF, Kahn M. Wnt signaling orchestration with a small molecule DYRK inhibitor provides long-term xeno-free human pluripotent cell expansion. Stem Cells Transl Med 2012; 1:18-28. [PMID: 23197636 PMCID: PMC3727690 DOI: 10.5966/sctm.2011-0033] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 10/26/2011] [Indexed: 12/31/2022] Open
Abstract
An optimal culture system for human pluripotent stem cells should be fully defined and free of animal components. To date, most xeno-free culture systems require human feeder cells and/or highly complicated culture media that contain activators of the fibroblast growth factor (FGF) and transforming growth factor-β (TGFβ) signaling pathways, and none provide for replacement of FGF/TGFβ ligands with chemical compounds. The Wnt/β-catenin signaling pathway plays an important role in mouse embryonic stem cells in leukemia inhibitory factor-independent culture; however, the role of Wnt/β-catenin signaling in human pluripotent stem cell is still poorly understood and controversial because of the dual role of Wnts in proliferation and differentiation. Building on our previous investigations of small molecules modulating Wnt/β-catenin signaling in mouse embryonic stem cells, we identified a compound, ID-8, that could support Wnt-induced human embryonic stem cell proliferation and survival without differentiation. Dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) is the target of the small molecule ID-8. Its role in human pluripotent cell renewal was confirmed by DYRK knockdown in human embryonic stem cells. Using Wnt and the DYRK inhibitor ID-8, we have developed a novel and simple chemically defined xeno-free culture system that allows for long-term expansion of human pluripotent stem cells without FGF or TGFβ activation. These culture conditions do not include xenobiotic supplements, serum, serum replacement, or albumin. Using this culture system, we have shown that several human pluripotent cell lines maintained pluripotency (>20 passages) and a normal karyotype and still retained the ability to differentiate into derivatives of all three germ layers. This Wnt-dependent culture system should provide a platform for complete replacement of growth factors with chemical compounds.
Collapse
Affiliation(s)
- Kouichi Hasegawa
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research
- Department of Cell and Neurobiology
- Institute for Integrated Cell-Material Sciences and
| | - Shin-ya Yasuda
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research
| | - Jia-Ling Teo
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research
| | - Cu Nguyen
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research
| | - Michael McMillan
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research
| | - Chih-Lin Hsieh
- Departments of Urology and Biochemistry and Molecular Biology, Norris Cancer Center
| | - Hirofumi Suemori
- Institute for Frontier Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Norio Nakatsuji
- Institute for Integrated Cell-Material Sciences and
- Institute for Frontier Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Masashi Yamamoto
- Functional Additives Division, Asahi Kasei Chemicals Corporation, Kawasaki, Kanagawa, Japan
| | | | - Carolyn Lutzko
- Department of Pediatrics, University of Southern California Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Martin F. Pera
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research
- Department of Cell and Neurobiology
| | - Michael Kahn
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research
- Department of Biochemistry and Molecular Biology, Keck School of Medicine
- Department of Molecular Pharmacology and Toxicology, and
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
1024
|
Mezzacappa C, Komiya Y, Habas R. Activation and function of small GTPases Rho, Rac, and Cdc42 during gastrulation. Methods Mol Biol 2012; 839:119-31. [PMID: 22218897 DOI: 10.1007/978-1-61779-510-7_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Gastrulation is comprised of a series of cell polarization and directional cell migration events that establish the physical body plan of the embryo. One of the major ligand-based pathways that has emerged to play crucial roles in the regulation of gastrulation is the non-canonical Wnt signaling pathway. This aspect of Wnt signaling is comprised of a number of signaling branches that are subsequently integrated for the regulation of changes to the actin cytoskeleton during cell polarization and cell migration during vertebrate gastrulation. The Rho family of small GTPases are activated and required for non-canonical Wnt signaling during gastrulation, and in this chapter, we describe biochemical assays for the detection of Wnt-mediated activation of Rho, Rac, and Cdc42 in both mammalian cells and Xenopus embryo explants.
Collapse
Affiliation(s)
- Courtney Mezzacappa
- Department of Biochemistry, UMDNJ-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | | | | |
Collapse
|
1025
|
Thorfve A, Dehne T, Lindahl A, Brittberg M, Pruss A, Ringe J, Sittinger M, Karlsson C. Characteristic Markers of the WNT Signaling Pathways Are Differentially Expressed in Osteoarthritic Cartilage. Cartilage 2012; 3:43-57. [PMID: 26069618 PMCID: PMC4297187 DOI: 10.1177/1947603511414178] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE It is well known that expression of markers for WNT signaling is dysregulated in osteoarthritic (OA) bone. However, it is still not fully known if the expression of these markers also is affected in OA cartilage. The aim of this study was therefore to examine this issue. METHODS Human cartilage biopsies from OA and control donors were subjected to genome-wide oligonucleotide microarrays. Genes involved in WNT signaling were selected using the BioRetis database, KEGG pathway analysis was searched using DAVID software tools, and cluster analysis was performed using Genesis software. Results from the microarray analysis were verified using quantitative real-time PCR and immunohistochemistry. In order to study the impact of cytokines for the dysregulated WNT signaling, OA and control chondrocytes were stimulated with interleukin-1 and analyzed with real-time PCR for their expression of WNT-related genes. RESULTS Several WNT markers displayed a significantly altered expression in OA compared to normal cartilage. Interestingly, inhibitors of the canonical and planar cell polarity WNT signaling pathways displayed significantly increased expression in OA cartilage, while the Ca(2+)/WNT signaling pathway was activated. Both real-time PCR and immunohistochemistry verified the microarray results. Real-time PCR analysis demonstrated that interleukin-1 upregulated expression of important WNT markers. CONCLUSIONS WNT signaling is significantly affected in OA cartilage. The result suggests that both the canonical and planar cell polarity WNT signaling pathways were partly inhibited while the Ca(2+)/WNT pathway was activated in OA cartilage.
Collapse
Affiliation(s)
- A. Thorfve
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Laboratory Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden,BIOMATCELL VINN Excellence Center of Biomaterials and Cell Therapy, Gothenburg, Sweden
| | - T. Dehne
- Department of Rheumatology and Clinical Immunology, Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - A. Lindahl
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Laboratory Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden,BIOMATCELL VINN Excellence Center of Biomaterials and Cell Therapy, Gothenburg, Sweden
| | - M. Brittberg
- Department of Orthopaedics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - A. Pruss
- Institute of Transfusion Medicine, Tissue Bank, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - J. Ringe
- Department of Rheumatology and Clinical Immunology, Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - M. Sittinger
- Department of Rheumatology and Clinical Immunology, Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - C. Karlsson
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Laboratory Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden,BIOMATCELL VINN Excellence Center of Biomaterials and Cell Therapy, Gothenburg, Sweden
| |
Collapse
|
1026
|
Fernández-Martos CM, González-Fernández C, González P, Maqueda A, Arenas E, Rodríguez FJ. Differential expression of Wnts after spinal cord contusion injury in adult rats. PLoS One 2011; 6:e27000. [PMID: 22073235 PMCID: PMC3206916 DOI: 10.1371/journal.pone.0027000] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 10/07/2011] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Spinal cord injury is a major cause of disability that has no clinically accepted treatment. Functional decline following spinal cord injury is caused by mechanical damage, secondary cell death, reactive gliosis and a poor regenerative capacity of damaged axons. Wnt proteins are a family of secreted glycoproteins that play key roles in different developmental processes although little is known of the expression patterns and functions of Wnts in the adult central nervous system in normal or diseased states. FINDINGS Using qRT-PCR analysis, we demonstrate that mRNA encoding most Wnt ligands and soluble inhibitors are constitutively expressed in the healthy adult spinal cord. Strikingly, contusion spinal cord injury induced a time-dependent increase in Wnt mRNA expression from 6 hours until 28 days post-injury, and a narrow peak in the expression of soluble Wnt inhibitors between 1 and 3 days post-injury. These results are consistent with the increase in the migration shift, from day 1 to 7, of the intracellular Wnt signalling component, Dishevelled-3. Moreover, after an initial decrease by 1 day, we also found an increase in phosphorylation of the Wnt co-receptor, low-density lipoprotein receptor-related protein 6, and an increase in active β-catenin protein, both of which suffer a dramatic change, from a homogeneous expression pattern in the grey matter to a disorganized injury-induced pattern. CONCLUSIONS Our results suggest a role for Wnts in spinal cord homeostasis and injury. We demonstrate that after injury Wnt signalling is activated via the Wnt/β-catenin and possibly other pathways. These findings provide an important foundation to further address the function of individual Wnt proteins in vivo and the pathophysiology of spinal cord injury.
Collapse
Affiliation(s)
| | | | - Pau González
- Laboratorio de Neurología Molecular, Hospital Nacional de Parapléjicos (HNP), Toledo, Spain
| | - Alfredo Maqueda
- Laboratorio de Neurología Molecular, Hospital Nacional de Parapléjicos (HNP), Toledo, Spain
| | - Ernest Arenas
- Molecular Neurobiology Unit, MBB, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
1027
|
Wnt/β-catenin pathway forms a negative feedback loop during TGF-β1 induced human normal skin fibroblast-to-myofibroblast transition. J Dermatol Sci 2011; 65:38-49. [PMID: 22041457 DOI: 10.1016/j.jdermsci.2011.09.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 07/17/2011] [Accepted: 09/30/2011] [Indexed: 01/12/2023]
Abstract
BACKGROUND Fibroblast-to-myofibroblast transition is a key event during wound healing and hypertrophic scar formation. Previous studies suggested Wnt/β-catenin signaling might be involved in the wound healing. However, its specific role in skin fibroblast-to-myofibroblast transition remains unclear. OBJECTIVE To investigate the specific role of β-catenin during the transforming growth factor-β1 induced normal skin myofibroblasts transition. METHODS By real-time quantitative polymerase chain reaction, Western-blot and immunocytochemistry, the activation of Wnt/β-catenin pathway in cultured human normal skin fibroblasts during TGF-β1 induced fibroblast-to-myofibroblast transition was investigated. The effects of β-catenin on myofibroblasts transition were also investigated when SB-216763, over-expression and siRNA of β-catenin were utilized. In addition, fibroblasts populated collagen lattices contraction assays were conducted to examine the effects of β-catenin on the contractility of the fibroblasts induced by TGF-β1. Furthermore, the effects of β-catenin on the expression of α-smooth muscle actin and collagen types I and III in hypertrophic scar derived fibroblasts were studied. RESULTS The expression of Wnts mRNA and β-catenin protein was up-regulated by TGF-β1 stimulation during the myofibroblasts transition. Both of SB-216763 and β-catenin over-expression was paralleled with decreased expression of α-smooth muscle actin, collagen types I and III, while siRNA targeting β-catenin leads to up-regulation of α-smooth muscle actin, collagen types I and III. The increased contractility and α-smooth muscle actin expression of the fibroblasts in the collagen lattices induced by TGF-β1 was inhibited by SB-216763. In addition, the expression levels of α-smooth muscle actin, collagen types I and III in hypertrophic scar derived fibroblasts were also down-regulated by SB-216763. CONCLUSION Specifically in normal skin fibroblasts, β-catenin might be involved in the myofibroblasts transition and negatively regulate the TGF-β1-induced myofibroblast transition.
Collapse
|
1028
|
Zheng F, Li H, Du W, Huang S. Role of hERG1 K(+) channels in leukemia cells as a positive regulator in SDF-1a-induced proliferation. ACTA ACUST UNITED AC 2011; 16:177-84. [PMID: 21669058 DOI: 10.1179/102453311x12940641878000] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Previous work from our laboratory has confirmed that human ether-à-go-go-related gene 1 (hERG1) K(+) channels are constitutively expressed in leukemia cells and enhanced cell proliferation. More importantly, it has shown that stromal cell-derived factor-1a (SDF-1a) significantly increases hERG1 K(+) tail current and a specific hERG1 K(+) channels inhibitor significantly blocks SDF-1a-induced migration of leukemic cells. In this study, we investigated a possible regulatory effect of hERG1 K(+) channels upon SDF-1a-mediated cell proliferation as a mean to uncover new molecular events involved in bone marrow microenvironment and leukemogenesis. RT-PCR showed that SDF-1a enhanced hERG1 expression in a dose-dependent manner. Cell proliferation assay illustrated that SDF-1a promoted cell proliferation in a dose-dependent manner, whereas this effect was impaired by E-4031. In addition, E-4031 inhibited SDF-1a-stimulated leukemic cell proliferation by inducing G(0)/G(1) arrest. Interestingly, E-4031 promoted SDF-1a-induced apoptosis in HL-60 and leukemic blasts, which markedly impaired the protection effect of SDF-1a in AML. Moreover, SDF-1a increased the expression of Wnt/beta-catenin target genes, including beta-catenin, cyclin-D1, and c-myc; however, this manner was abolished by blockage with the hERG1 K(+) channels. Taken together, our results provide evidence of a novel mechanism involved in the proliferative effects of SDF-1a and highlight hERG1 K(+) channels as a therapeutic target for leukemia treatment and prevention.
Collapse
Affiliation(s)
- Fang Zheng
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | |
Collapse
|
1029
|
Quantitative and kinetic profile of Wnt/β-catenin signaling components during human neural progenitor cell differentiation. Cell Mol Biol Lett 2011; 16:515-38. [PMID: 21805133 PMCID: PMC6275579 DOI: 10.2478/s11658-011-0021-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 08/03/2011] [Indexed: 12/30/2022] Open
Abstract
ReNcell VM is an immortalized human neural progenitor cell line with the ability to differentiate in vitro into astrocytes and neurons, in which the Wnt/β-catenin pathway is known to be involved. However, little is known about kinetic changes of this pathway in human neural progenitor cell differentiation. In the present study, we provide a quantitative profile of Wnt/β-catenin pathway dynamics showing its spatio-temporal regulation during ReNcell VM cell differentiation. We show first that T-cell factor dependent transcription can be activated by stabilized β-catenin. Furthermore, endogenous Wnt ligands, pathway receptors and signaling molecules are temporally controlled, demonstrating changes related to differentiation stages. During the first three hours of differentiation the signaling molecules LRP6, Dvl2 and β-catenin are spatio-temporally regulated between distinct cellular compartments. From 24 h onward, components of the Wnt/β-catenin pathway are strongly activated and regulated as shown by mRNA up-regulation of Wnt ligands (Wnt5a and Wnt7a), receptors including Frizzled-2, -3, -6, -7, and -9, and co-receptors, and target genes including Axin2. This detailed temporal profile of the Wnt/β-catenin pathway is a first step to understand, control and to orientate, in vitro, human neural progenitor cell differentiation.
Collapse
|
1030
|
Miller RK, Canny SGDLT, Jang CW, Cho K, Ji H, Wagner DS, Jones EA, Habas R, McCrea PD. Pronephric tubulogenesis requires Daam1-mediated planar cell polarity signaling. J Am Soc Nephrol 2011; 22:1654-64. [PMID: 21804089 DOI: 10.1681/asn.2010101086] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Canonical β-catenin-mediated Wnt signaling is essential for the induction of nephron development. Noncanonical Wnt/planar cell polarity (PCP) pathways contribute to processes such as cell polarization and cytoskeletal modulation in several tissues. Although PCP components likely establish the plane of polarization in kidney tubulogenesis, whether PCP effectors directly modulate the actin cytoskeleton in tubulogenesis is unknown. Here, we investigated the roles of Wnt PCP components in cytoskeletal assembly during kidney tubule morphogenesis in Xenopus laevis and zebrafish. We found that during tubulogenesis, the developing pronephric anlagen expresses Daam1 and its interacting Rho-GEF (WGEF), which compose one PCP/noncanonical Wnt pathway branch. Knockdown of Daam1 resulted in reduced expression of late pronephric epithelial markers with no apparent effect upon early markers of patterning and determination. Inhibiting various points in the Daam1 signaling pathway significantly reduced pronephric tubulogenesis. These data indicate that pronephric tubulogenesis requires the Daam1/WGEF/Rho PCP pathway.
Collapse
Affiliation(s)
- Rachel K Miller
- Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1031
|
Potential cross-talk between (pro)renin receptors and Wnt/frizzled receptors in cardiovascular and renal disorders. Hypertens Res 2011; 34:1161-70. [DOI: 10.1038/hr.2011.113] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
1032
|
Miller MM, Iglesias DM, Zhang Z, Corsini R, Chu L, Murawski I, Gupta I, Somlo S, Germino GG, Goodyer PR. T-cell factor/β-catenin activity is suppressed in two different models of autosomal dominant polycystic kidney disease. Kidney Int 2011; 80:146-53. [PMID: 21389971 PMCID: PMC3879791 DOI: 10.1038/ki.2011.56] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During murine kidney development, canonical WNT signaling is highly active in tubules until about embryonic days E16-E18. At this time, β-catenin transcriptional activity is progressively restricted to the nephrogenic zone. The cilial protein genes PKD1 and PKD2 are known to be mutated in autosomal dominant polycystic kidney disease (ADPKD), and previous studies proposed that these mutations could lead to a failure to suppress canonical WNT signaling activity. Several in vitro studies have found a link between cilial signaling and β-catenin regulation, suggesting that aberrant activity might contribute to the cystic phenotype. To study this, we crossed T-cell factor (TCF)/β-catenin-lacZ reporter mice with mice having Pkd1 or Pkd2 mutations and found that there was no β-galactosidase staining in cells lining the renal cysts. Thus, suppression of canonical WNT activity, defined by the TCF/β-catenin-lacZ reporter, is normal in these two different models of polycystic kidney disease. Hence, excessive β-catenin transcriptional activity may not contribute to cystogenesis in these models of ADPKD.
Collapse
Affiliation(s)
- Michelle M. Miller
- Department of Human Genetics, McGill University-Montreal Children’s Hospital Research Institute, McGill University Health Centre Research Institute and McGill University, Montreal, Quebec, Canada
| | - Diana M. Iglesias
- Department of Pediatrics, McGill University-Montreal Children’s Hospital Research Institute, McGill University Health Centre Research Institute and McGill University, Montreal, Quebec, Canada
| | - Zhao Zhang
- Department of Pediatrics, McGill University-Montreal Children’s Hospital Research Institute, McGill University Health Centre Research Institute and McGill University, Montreal, Quebec, Canada
| | - Rachel Corsini
- Department of Pediatrics, McGill University-Montreal Children’s Hospital Research Institute, McGill University Health Centre Research Institute and McGill University, Montreal, Quebec, Canada
| | - LeeLee Chu
- Department of Pediatrics, McGill University-Montreal Children’s Hospital Research Institute, McGill University Health Centre Research Institute and McGill University, Montreal, Quebec, Canada
| | - Inga Murawski
- Department of Human Genetics, McGill University-Montreal Children’s Hospital Research Institute, McGill University Health Centre Research Institute and McGill University, Montreal, Quebec, Canada
| | - Indra Gupta
- Department of Pediatrics, McGill University-Montreal Children’s Hospital Research Institute, McGill University Health Centre Research Institute and McGill University, Montreal, Quebec, Canada
| | - Stefan Somlo
- Department of Genetics and Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Gregory G. Germino
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Paul R. Goodyer
- Department of Human Genetics, McGill University-Montreal Children’s Hospital Research Institute, McGill University Health Centre Research Institute and McGill University, Montreal, Quebec, Canada
- Department of Pediatrics, McGill University-Montreal Children’s Hospital Research Institute, McGill University Health Centre Research Institute and McGill University, Montreal, Quebec, Canada
| |
Collapse
|
1033
|
Okerlund ND, Cheyette BNR. Synaptic Wnt signaling-a contributor to major psychiatric disorders? J Neurodev Disord 2011; 3:162-74. [PMID: 21533542 PMCID: PMC3180925 DOI: 10.1007/s11689-011-9083-6] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 04/05/2011] [Indexed: 02/08/2023] Open
Abstract
Wnt signaling is a key pathway that helps organize development of the nervous system. It influences cell proliferation, cell fate, and cell migration in the developing nervous system, as well as axon guidance, dendrite development, and synapse formation. Given this wide range of roles, dysregulation of Wnt signaling could have any number of deleterious effects on neural development and thereby contribute in many different ways to the pathogenesis of neurodevelopmental disorders. Some major psychiatric disorders, including schizophrenia, bipolar disorder, and autism spectrum disorders, are coming to be understood as subtle dysregulations of nervous system development, particularly of synapse formation and maintenance. This review will therefore touch on the importance of Wnt signaling to neurodevelopment generally, while focusing on accumulating evidence for a synaptic role of Wnt signaling. These observations will be discussed in the context of current understanding of the neurodevelopmental bases of major psychiatric diseases, spotlighting schizophrenia, bipolar disorder, and autism spectrum disorder. In short, this review will focus on the potential role of synapse formation and maintenance in major psychiatric disorders and summarize evidence that defective Wnt signaling could contribute to their pathogenesis via effects on these late neural differentiation processes.
Collapse
Affiliation(s)
- Nathan D. Okerlund
- Neuroscience Program, University of California, San Francisco, 1550 4th St, San Francisco, CA 94158 USA
- Nina Ireland Laboratory for Developmental Neurobiology, Department of Psychiatry, University of California, San Francisco, 1550 4th St, San Francisco, CA 94158 USA
| | - Benjamin N. R. Cheyette
- Neuroscience Program, University of California, San Francisco, 1550 4th St, San Francisco, CA 94158 USA
- Program in Developmental and Stem Cell Biology, University of California, San Francisco, 1550 4th St, San Francisco, CA 94158 USA
- Nina Ireland Laboratory for Developmental Neurobiology, Department of Psychiatry, University of California, San Francisco, 1550 4th St, San Francisco, CA 94158 USA
| |
Collapse
|
1034
|
Yang L, Wu X, Wang Y, Zhang K, Wu J, Yuan YC, Deng X, Chen L, Kim CCH, Lau S, Somlo G, Yen Y. FZD7 has a critical role in cell proliferation in triple negative breast cancer. Oncogene 2011; 30:4437-46. [PMID: 21532620 DOI: 10.1038/onc.2011.145] [Citation(s) in RCA: 220] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Breast cancer is genetically and clinically heterogeneous. Triple negative breast cancer (TNBC) is a subtype of breast cancer that is usually associated with poor outcome and lack of benefit from targeted therapy. We used microarray analysis to perform a pathway analysis of TNBC compared with non-triple negative breast cancer (non-TNBC). Overexpression of several Wnt pathway genes, such as frizzled homolog 7 (FZD7), low density lipoprotein receptor-related protein 6 and transcription factor 7 (TCF7) was observed in TNBC, and we directed our focus to the Wnt pathway receptor, FZD7. To validate the function of FZD7, FZD7shRNA was used to knock down FZD7 expression. Notably, reduced cell proliferation and suppressed invasiveness and colony formation were observed in TNBC MDA-MB-231 and BT-20 cells. Study of the possible mechanism indicated that these effects occurred through silencing of the canonical Wnt signaling pathway, as evidenced by loss of nuclear accumulation of β-catenin and decreased transcriptional activity of TCF7. In vivo studies revealed that FZD7shRNA significantly suppressed tumor formation, through reduced cell proliferation, in mice bearing xenografts without FZD7 expression. Our findings suggest that FZD7-involved canonical Wnt signaling pathway is essential for tumorigenesis of TNBC, and thus, FZD7 shows promise as a biomarker and a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- L Yang
- Department of Molecular Pharmacology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1035
|
Bahls M, Bidwell CA, Hu J, Krueger CG, Reed JD, Tellez A, Kaluza GL, Granada JF, Van Alstine WG, Newcomer SC. Gene expression differences in healthy brachial and femoral arteries of Rapacz familial hypercholesterolemic swine. Physiol Genomics 2011; 43:781-8. [PMID: 21505098 DOI: 10.1152/physiolgenomics.00151.2010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The mechanisms underlying the unequal distribution of atherosclerotic disease in the peripheral arteries are currently unclear. Gene expression differences in healthy arteries may influence the heterogeneous distribution of atherosclerosis. Therefore, this investigation compares gene expression in healthy atheroprotected brachial and atherosusceptible femoral arteries of young and disease free Rapacz familial hypercholesterolemic (FHC) swine. We hypothesized that transcripts related to atherosusceptibility would be differentially expressed between these arteries prior to the onset of disease. Femoral and brachial arteries were harvested from four 13-day-old Rapacz FHC swine. No atherosclerotic disease was detected using Sudan IV, Verhoeff-van Gieson, and hematoxylin-eosin staining. Gene expression was quantified using Affymetrix GeneChip Porcine Genome Arrays. An average of 15,552 probe sets had detectable transcripts, while 430 probe sets showed a significant differential expression between arteries (false discovery rate < 0.05). The human orthologs of 63 probe sets with differential expression and a 1.5-fold or greater transcript abundance between arteries are associated with Wnt/β-catenin, lysophospholipid, and Ca-signaling, as well as apoptosis. This is the first investigation reporting that differences in relative abundance of gene expression exist between brachial and femoral arteries in young Rapacz FHC swine prior to the development of atherosclerotic lesions.
Collapse
Affiliation(s)
- Martin Bahls
- Departments of Health and Kinesiology, Purdue University, West Lafayette, Indiana, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1036
|
Yao H, Ashihara E, Maekawa T. Targeting the Wnt/β-catenin signaling pathway in human cancers. Expert Opin Ther Targets 2011; 15:873-87. [PMID: 21486121 DOI: 10.1517/14728222.2011.577418] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The Wnt/β-catenin signaling pathway plays a pivotal role in the regulation of cell growth, cell development and the differentiation of normal stem cells. Constitutive activation of the Wnt/β-catenin signaling pathway is found in many human cancers, and is thus an attractive target for anti-cancer therapy. Specific inhibitors of this pathway have been keenly researched and developed. AREAS COVERED This review discusses the potential of inhibiting the Wnt/β-catenin signaling pathway, as a therapeutic approach for cancer, along with an overview of the development of specific inhibitors. EXPERT OPINION Cancer stem cells (CSCs) play a significant role in the development and recurrence of several cancers, and Wnt/β-catenin signaling is important for the proliferation of CSCs. Inhibition of Wnt/β-catenin signaling is therefore a promising treatment approach. Progress has been made in the development of screening methods to identify Wnt/β-catenin signaling inhibitors. Biomarker-based screening is an effective and promising method for the identification of compounds of interest.
Collapse
Affiliation(s)
- Hisayuki Yao
- Kyoto University Hospital, Department of Transfusion Medicine and Cell Therapy, Kyoto, Japan
| | | | | |
Collapse
|
1037
|
Vangamudi B, Zhu S, Soutto M, Belkhiri A, El-Rifai W. Regulation of β-catenin by t-DARPP in upper gastrointestinal cancer cells. Mol Cancer 2011; 10:32. [PMID: 21447180 PMCID: PMC3075216 DOI: 10.1186/1476-4598-10-32] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 03/29/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Truncated dopamine and cyclic-AMP-regulated phosphoprotein (t-DARPP) is frequently overexpressed in gastrointestinal malignancies. In this study, we examined the role of t-DARPP in regulating β-catenin. RESULTS The pTopFlash construct that contains multiple TCF/LEF-binding sites was used as a measure of β-catenin/TCF transcription activity. Gastric (AGS, MKN28) and esophageal (FLO-1) adenocarcinoma cancer cell lines that lack t-DARPP expression were utilized to establish stable and transient in vitro expression models of t-DARPP. The expression of t-DARPP led to a significant induction of the pTOP reporter activity, indicative of activation of β-catenin/TCF nuclear signaling. Immunofluorescence assays supported this finding and showed accumulation and nuclear translocation of β-catenin in cells expressing t-DARPP. These cells had a significant increase in their proliferative capacity and demonstrated up-regulation of two transcription targets of β-catenin/TCF: Cyclin D1 and c-MYC. Because phosphorylated GSK-3β is inactive and loses its ability to phosphorylate β-catenin and target it towards degradation by the proteasome, we next examined the levels of phospho-GSK-3β. These results demonstrated an increase in phospho-GSK-3β and phospho-AKT. The knockdown of endogenous t-DARPP in MKN45 cancer cells demonstrated a reversal of the signaling events. To examine whether t-DARPP mediated GSK-3β phosphorylation in an AKT-dependent manner, we used a pharmacologic inhibitor of PI3K/AKT, LY294002, in cancer cells expressing t-DARPP. This treatment abolished the phosphorylation of AKT and GSK-3β leading to a reduction in β-catenin, Cyclin D1, and c-MYC protein levels. CONCLUSIONS Our findings demonstrate, for the first time, that t-DARPP regulates β-catenin/TCF activity, thereby implicating a novel oncogenic signaling in upper gastrointestinal cancers.
Collapse
Affiliation(s)
- Bhavatarini Vangamudi
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Shoumin Zhu
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mohammed Soutto
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Abbes Belkhiri
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wael El-Rifai
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
1038
|
Modulation of cell adhesion systems by prenatal nicotine exposure in limbic brain regions of adolescent female rats. Int J Neuropsychopharmacol 2011; 14:157-74. [PMID: 20196919 PMCID: PMC5575906 DOI: 10.1017/s1461145710000179] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Maternal smoking during pregnancy (MS) has long-lasting neurobehavioural effects on the offspring. Many MS-associated psychiatric disorders begin or change symptomatology during adolescence, a period of continuous development of the central nervous system. However, the underlying molecular mechanisms are largely unknown. Given that cell adhesion molecules (CAMs) modulate various neurotransmitter systems and are associated with many psychiatric disorders, we hypothesize that CAMs are altered by prenatal treatment of nicotine, the major psychoactive component in tobacco, in adolescent brains. Pregnant Sprague-Dawley rats were treated with nicotine (3 mg/kg.d) or saline via osmotic mini-pumps from gestational days 4 to 18. Female offspring at postnatal day 35 were sacrificed, and several limbic brain regions (the caudate putamen, nucleus accumbens, prefrontal cortex, and amygdala) were dissected for evaluation of gene expression using microarray and quantitative RT-PCR techniques. Various CAMs including neurexin, immunoglobulin, cadherin, and adhesion-GPCR superfamilies, and their intracellular signalling pathways were modified by gestational nicotine treatment (GN). Among the CAM-related pathways, GN has stronger effects on cytoskeleton reorganization pathways than on gene transcription pathways. These effects were highly region dependent, with the caudate putamen showing the greatest vulnerability. Given the important roles of CAMs in neuronal development and synaptic plasticity, our findings suggest that alteration of CAMs contributes to the neurobehavioural deficits associated with MS. Further, our study underscores that low doses of nicotine produce substantial and long-lasting changes in the brain, implying that nicotine replacement therapy during pregnancy may carry many of the same risks to the offspring as MS.
Collapse
|
1039
|
Liu W, Su LT, Khadka DK, Mezzacappa C, Komiya Y, Sato A, Habas R, Runnels LW. TRPM7 regulates gastrulation during vertebrate embryogenesis. Dev Biol 2011; 350:348-57. [PMID: 21145885 PMCID: PMC3292586 DOI: 10.1016/j.ydbio.2010.11.034] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 11/30/2010] [Accepted: 11/30/2010] [Indexed: 02/06/2023]
Abstract
During gastrulation, cells in the dorsal marginal zone polarize, elongate, align and intercalate to establish the physical body axis of the developing embryo. Here we demonstrate that the bifunctional channel-kinase TRPM7 is specifically required for vertebrate gastrulation. TRPM7 is temporally expressed maternally and throughout development, and is spatially enriched in tissues undergoing convergent extension during gastrulation. Functional studies reveal that TRPM7's ion channel, but not its kinase domain, specifically affects cell polarity and convergent extension movements during gastrulation, independent of mesodermal specification. During gastrulation, the non-canonical Wnt pathway via Dishevelled (Dvl) orchestrates the activities of the GTPases Rho and Rac to control convergent extension movements. We find that TRPM7 functions synergistically with non-canonical Wnt signaling to regulate Rac activity. The phenotype caused by depletion of the Ca(2+)- and Mg(2+)-permeant TRPM7 is suppressed by expression of a dominant negative form of Rac, as well as by Mg(2+) supplementation or by expression of the Mg(2+) transporter SLC41A2. Together, these studies demonstrate an essential role for the ion channel TRPM7 and Mg(2+) in Rac-dependent polarized cell movements during vertebrate gastrulation.
Collapse
Affiliation(s)
- Wei Liu
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | | | | | | | | | | | | | | |
Collapse
|
1040
|
Dishevelled is essential for neural connectivity and planar cell polarity in planarians. Proc Natl Acad Sci U S A 2011; 108:2813-8. [PMID: 21282632 DOI: 10.1073/pnas.1012090108] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Wingless/Integrated (Wnt) signaling pathway controls multiple events during development and homeostasis. It comprises multiple branches, mainly classified according to their dependence on β-catenin activation. The Wnt/β-catenin branch is essential for the establishment of the embryonic anteroposterior (AP) body axis throughout the phylogenetic tree. It is also required for AP axis establishment during planarian regeneration. Wnt/β-catenin-independent signaling encompasses several different pathways, of which the most extensively studied is the planar cell polarity (PCP) pathway, which is responsible for planar polarization of cell structures within an epithelial sheet. Dishevelled (Dvl) is the hub of Wnt signaling because it regulates and channels the Wnt signal into every branch. Here, we analyze the role of Schmidtea mediterranea Dvl homologs (Smed-dvl-1 and Smed-dvl-2) using gene silencing. We demonstrate that in addition to a role in AP axis specification, planarian Dvls are involved in at least two different β-catenin-independent processes. First, they are essential for neural connectivity through Smed-wnt5 signaling. Second, Smed-dvl-2, together with the S. mediterranea homologs of Van-Gogh (Vang) and Diversin (Div), is required for apical positioning of the basal bodies of epithelial cells. These data represent evidence not only of the function of the PCP network in lophotrocozoans but of the involvement of the PCP core elements Vang and Div in apical positioning of the cilia.
Collapse
|
1041
|
Mathew SJ, Hansen JM, Merrell AJ, Murphy MM, Lawson JA, Hutcheson DA, Hansen MS, Angus-Hill M, Kardon G. Connective tissue fibroblasts and Tcf4 regulate myogenesis. Development 2011; 138:371-84. [PMID: 21177349 DOI: 10.1242/dev.057463] [Citation(s) in RCA: 244] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Muscle and its connective tissue are intimately linked in the embryo and in the adult, suggesting that interactions between these tissues are crucial for their development. However, the study of muscle connective tissue has been hindered by the lack of molecular markers and genetic reagents to label connective tissue fibroblasts. Here, we show that the transcription factor Tcf4 (transcription factor 7-like 2; Tcf7l2) is strongly expressed in connective tissue fibroblasts and that Tcf4(GFPCre) mice allow genetic manipulation of these fibroblasts. Using this new reagent, we find that connective tissue fibroblasts critically regulate two aspects of myogenesis: muscle fiber type development and maturation. Fibroblasts promote (via Tcf4-dependent signals) slow myogenesis by stimulating the expression of slow myosin heavy chain. Also, fibroblasts promote the switch from fetal to adult muscle by repressing (via Tcf4-dependent signals) the expression of developmental embryonic myosin and promoting (via a Tcf4-independent mechanism) the formation of large multinucleate myofibers. In addition, our analysis of Tcf4 function unexpectedly reveals a novel mechanism of intrinsic regulation of muscle fiber type development. Unlike other intrinsic regulators of fiber type, low levels of Tcf4 in myogenic cells promote both slow and fast myogenesis, thereby promoting overall maturation of muscle fiber type. Thus, we have identified novel extrinsic and intrinsic mechanisms regulating myogenesis. Most significantly, our data demonstrate for the first time that connective tissue is important not only for adult muscle structure and function, but is a vital component of the niche within which muscle progenitors reside and is a critical regulator of myogenesis.
Collapse
Affiliation(s)
- Sam J Mathew
- Department of Human Genetics, University of Utah, 15 North 2030 East, Salt Lake City, Utah 84112, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1042
|
Dishevelled-3 C-terminal His single amino acid repeats are obligate for Wnt5a activation of non-canonical signaling. J Mol Signal 2010; 5:19. [PMID: 21092292 PMCID: PMC3003240 DOI: 10.1186/1750-2187-5-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 11/23/2010] [Indexed: 11/25/2022] Open
Abstract
Background The Wnt non-canonical pathway (Wnt5a > Frizzled-2 > cyclic GMP phosphodiesterase/Ca2+-mobilization pathway regulates the activation of NF-AT) is mediated by three mammalian Dishevelleds (Dvl1, Dvl2, and Dvl3) and the role of the C-terminal region unique to Dvl3 was interrogated. Results Dvl1, Dvl2, and Dvl3 are expressed at varying levels in mouse totipotent F9 embryonal teratocarcinoma cells. The expression of each endogenous Dvl isoform, as defined by knock-down with siRNA, was obligate for Wnt5a to activate NF-AT-sensitive transcription. Elements upstream of effectors, e.g., cGMP phosphodiesterase and Ca2+-mobilization, were blocked by knock-down of any one of the Dvls; thus, with respect to Wnt5a activation of NF-AT Dvls are not redundant. Among the three Dvl isoforms, the C-terminal sequence of Dvl3 is the most divergent. Deletion of region of Dvl3 abolishes Wnt5a-stimulated signaling. Alanine (Ala)-substitution of histidine (His) single amino acid repeats at 637,638 and/or 647,648 in Dvl3, like C-terminal deletion, abolishes Wnt 5a signal propagation. Phenylalanine (Phe)-substitution of the same His-repeats in Dvl3 mimics Wnt5a stimulated NF-AT-sensitive transcription. Conclusions The C-terminal third of Dvl3 and His single amino acid repeats 637,638 and 647,648 (which are unique to and conserved in Dvl3) are essential for Wnt5a activation of the non-canonical pathway, but not the Wnt3a activation of the canonical pathway.
Collapse
|
1043
|
Sonderegger S, Pollheimer J, Knöfler M. Wnt signalling in implantation, decidualisation and placental differentiation--review. Placenta 2010; 31:839-47. [PMID: 20716463 PMCID: PMC2963059 DOI: 10.1016/j.placenta.2010.07.011] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 07/20/2010] [Accepted: 07/21/2010] [Indexed: 12/19/2022]
Abstract
The family of secreted Wingless ligands plays major roles in embryonic development, stem cell maintenance, differentiation and tissue homeostasis. Accumulating evidence suggests that the canonical Wnt pathway involving nuclear recruitment of β-catenin and activation of Wnt-dependent transcription factors is also critically involved in development and differentiation of the diverse reproductive tissues. Here, we summarise our present knowledge about expression, regulation and function of Wnt ligands and their frizzled receptors in murine and human endometrial and placental cell types. In mice, Wnt signalling promotes early trophoblast lineage development, blastocyst activation, implantation and chorion-allantois fusion. Moreover, different Wnt ligands play essential roles in the development of the murine uterine tract, in cycling endometrial cells and during decidualisation. In humans, estrogen-dependent endometrial cell proliferation, decidualisation, trophoblast attachment and invasion were shown to be controlled by the particular signalling pathway. Failures in Wnt signalling are associated with infertility, endometriosis, endometrial cancer and gestational diseases such as complete mole placentae and choriocarcinomas. However, our present knowledge is still scarce due to the complexity of the Wnt network involving numerous ligands, receptors and non-canonical pathways. Hence, much remains to be learned about the role of different Wnt signalling cascades in reproductive cell types and their changes under pathological conditions.
Collapse
Affiliation(s)
- S. Sonderegger
- Prince Henry’s Institute of Medical Research, Clayton, Victoria, Australia
| | - J. Pollheimer
- Department of Obstetrics and Fetal–Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - M. Knöfler
- Department of Obstetrics and Fetal–Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| |
Collapse
|
1044
|
Rieger ME, Sims AH, Coats ER, Clarke RB, Briegel KJ. The embryonic transcription cofactor LBH is a direct target of the Wnt signaling pathway in epithelial development and in aggressive basal subtype breast cancers. Mol Cell Biol 2010; 30:4267-79. [PMID: 20606007 PMCID: PMC2937547 DOI: 10.1128/mcb.01418-09] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 12/29/2009] [Accepted: 06/22/2010] [Indexed: 12/11/2022] Open
Abstract
Limb-bud and heart (LBH) is a novel key transcriptional regulator of vertebrate development. However, the molecular mechanisms upstream of LBH and its role in adult development are unknown. Here we show that in epithelial development, LBH expression is tightly controlled by Wnt signaling. LBH is transcriptionally induced by the canonical Wnt pathway, as evident by the presence of conserved functional T-cell factor (TCF)/lymphoid enhancer-binding factor (LEF) binding sites in the LBH locus and rapid beta-catenin-dependent upregulation of endogenous LBH by Wnt3a. In contrast, LBH induction by Wnt/beta-catenin signaling is inhibited by Wnt7a, which in limb development signals through a noncanonical pathway involving Lmx1b. Furthermore, we show that LBH is aberrantly overexpressed in mammary tumors of mouse mammary tumor virus (MMTV)-Wnt1-transgenic mice and in aggressive basal subtype human breast cancers that display Wnt/beta-catenin hyperactivation. Deregulation of LBH in human basal breast cancer appears to be Wnt/beta-catenin dependent, as DKK1 and Wnt7a inhibit LBH expression in breast tumor cells. Overexpression studies indicate that LBH suppresses mammary epithelial cell differentiation, an effect that could contribute to Wnt-induced tumorigenesis. Taken together, our findings link LBH for the first time to the Wnt signaling pathway in both development and cancer and highlight LBH as a potential new marker for therapeutically challenging basal-like breast cancers.
Collapse
Affiliation(s)
- Megan E. Rieger
- Department of Biochemistry and Molecular Biology, Braman Family Breast Cancer Institute at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, Breast Biology Group, Paterson Institute for Cancer Research, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4BX, United Kingdom
| | - Andrew H. Sims
- Department of Biochemistry and Molecular Biology, Braman Family Breast Cancer Institute at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, Breast Biology Group, Paterson Institute for Cancer Research, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4BX, United Kingdom
| | - Ebony R. Coats
- Department of Biochemistry and Molecular Biology, Braman Family Breast Cancer Institute at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, Breast Biology Group, Paterson Institute for Cancer Research, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4BX, United Kingdom
| | - Robert B. Clarke
- Department of Biochemistry and Molecular Biology, Braman Family Breast Cancer Institute at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, Breast Biology Group, Paterson Institute for Cancer Research, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4BX, United Kingdom
| | - Karoline J. Briegel
- Department of Biochemistry and Molecular Biology, Braman Family Breast Cancer Institute at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, Breast Biology Group, Paterson Institute for Cancer Research, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4BX, United Kingdom
| |
Collapse
|
1045
|
Bulla GA, Luong Q, Shrestha S, Reeb S, Hickman S. Genome-wide analysis of hepatic gene silencing in mammalian cell hybrids. Genomics 2010; 96:323-32. [PMID: 20801210 DOI: 10.1016/j.ygeno.2010.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 08/13/2010] [Accepted: 08/17/2010] [Indexed: 12/29/2022]
Abstract
Silencing of tissue-specific gene expression in mammalian somatic cell hybrids is a well-documented epigenetic phenomenon which is both profound (involving a large number of genes) and enigmatic. Our aim was to utilize whole-genome microarray analyses to determine the true extent of gene silencing on a genomic level. By comparing gene expression profiles of hepatoma×fibroblast cell hybrids with those of parental cells, we have identified over 300 liver-enriched genes that are repressed at least 5-fold in the cell hybrids, the majority of which are repressed at least 10-fold. Also, we identify nearly 200 fibroblast-enriched genes that are repressed at least 5-fold. Silenced hepatic genes include several that encode transcription factors and proteins involved in signal transduction pathways. These data suggest that extensive reprogramming occurs in cell hybrids, leading to a nearly global (although not complete) loss of tissue-specific gene expression.
Collapse
Affiliation(s)
- Gary A Bulla
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston, IL 61920, USA.
| | | | | | | | | |
Collapse
|
1046
|
Abstract
The differentiation of embryonic stem cells along the endothelial cell lineage requires a tightly coordinated sequence of events that are regulated in both space and time. Although significant gaps remain in this process, major strides have been made over the past 10 years in identifying the growth factors, signal transduction pathways, and transcription factors that function together as critical mediators of this process. Examples of some of the signal transduction pathways include the hedgehog (HH), WNT, BMP, and Notch pathways. A complex interplay between growth factors, and activation of a variety of signal transduction pathways leads to the induction of transcriptional programs that promote the differentiation of embryonic stem cells along the endothelial lineage and ultimately into arterial, venous, and lymphatic endothelial cells. The purpose of this review is to summarize the recent advances in our understanding of the molecular mechanisms underlying endothelial differentiation.
Collapse
Affiliation(s)
- Alex Le Bras
- Division of Cardiology, and Molecular and Vascular Biology, Department of Medicine and the Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | | |
Collapse
|
1047
|
Liu Z, Habener JF. Wnt signaling in pancreatic islets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:391-419. [PMID: 20217507 DOI: 10.1007/978-90-481-3271-3_17] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The Wnt signaling pathway is critically important not only for stem cell amplification, differentiation, and migration, but also is important for organogenesis and the development of the body plan. Beta-catenin/TCF7L2-dependent Wnt signaling (the canonical pathway) is involved in pancreas development, islet function, and insulin production and secretion. The glucoincretin hormone glucagon-like peptide-1 and the chemokine stromal cell-derived factor-1 modulate canonical Wnt signaling in beta-cells which is obligatory for their mitogenic and cytoprotective actions. Genome-wide association studies have uncovered 19 gene loci that confer susceptibility for the development of type 2 diabetes. At least 14 of these diabetes risk alleles encode proteins that are implicated in islet growth and functioning. Seven of them are either components of, or known target genes for, Wnt signaling. The transcription factor TCF7L2 is particularly strongly associated with risk for diabetes and appears to be fundamentally important in both canonical Wnt signaling and beta-cell functioning. Experimental loss of TCF7L2 function in islets and polymorphisms in TCF7L2 alleles in humans impair glucose-stimulated insulin secretion, suggesting that perturbations in the Wnt signaling pathway may contribute substantially to the susceptibility for, and pathogenesis of, type 2 diabetes. This review focuses on considerations of the hormonal regulation of Wnt signaling in islets and implications for mutations in components of the Wnt signaling pathway as a source for risk-associated alleles for type 2 diabetes.
Collapse
Affiliation(s)
- Zhengyu Liu
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | |
Collapse
|
1048
|
Lukaszewicz AI, McMillan MK, Kahn M. Small molecules and stem cells. Potency and lineage commitment: the new quest for the fountain of youth. J Med Chem 2010; 53:3439-53. [PMID: 20047330 DOI: 10.1021/jm901361d] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Agnès I Lukaszewicz
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
| | | | | |
Collapse
|
1049
|
Yates LL, Schnatwinkel C, Murdoch JN, Bogani D, Formstone CJ, Townsend S, Greenfield A, Niswander LA, Dean CH. The PCP genes Celsr1 and Vangl2 are required for normal lung branching morphogenesis. Hum Mol Genet 2010; 19:2251-67. [PMID: 20223754 PMCID: PMC2865378 DOI: 10.1093/hmg/ddq104] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 03/08/2010] [Indexed: 12/24/2022] Open
Abstract
The lungs are generated by branching morphogenesis as a result of reciprocal signalling interactions between the epithelium and mesenchyme during development. Mutations that disrupt formation of either the correct number or shape of epithelial branches affect lung function. This, in turn, can lead to congenital abnormalities such as cystadenomatoid malformations, pulmonary hypertension or lung hypoplasia. Defects in lung architecture are also associated with adult lung disease, particularly in cases of idiopathic lung fibrosis. Identifying the signalling pathways which drive epithelial tube formation will likely shed light on both congenital and adult lung disease. Here we show that mutations in the planar cell polarity (PCP) genes Celsr1 and Vangl2 lead to disrupted lung development and defects in lung architecture. Lungs from Celsr1(Crsh) and Vangl2(Lp) mouse mutants are small and misshapen with fewer branches, and by late gestation exhibit thickened interstitial mesenchyme and defective saccular formation. We observe a recapitulation of these branching defects following inhibition of Rho kinase, an important downstream effector of the PCP signalling pathway. Moreover, epithelial integrity is disrupted, cytoskeletal remodelling perturbed and mutant endoderm does not branch normally in response to the chemoattractant FGF10. We further show that Celsr1 and Vangl2 proteins are present in restricted spatial domains within lung epithelium. Our data show that the PCP genes Celsr1 and Vangl2 are required for foetal lung development thereby revealing a novel signalling pathway critical for this process that will enhance our understanding of congenital and adult lung diseases and may in future lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- Laura L. Yates
- Medical Research Council, Harwell, Oxfordshire OX11 0RD, UK
| | - Carsten Schnatwinkel
- Howard Hughes Medical Institute, Department of Pediatrics, Section of Developmental Biology, University of Colorado Denver School of Medicine, Aurora, CO, USA and
| | | | - Debora Bogani
- Medical Research Council, Harwell, Oxfordshire OX11 0RD, UK
| | - Caroline J. Formstone
- MRC Centre for Developmental Neurobiology, Kings College London, New Hunts House, London SE1 1UL, UK
| | | | | | - Lee A. Niswander
- Howard Hughes Medical Institute, Department of Pediatrics, Section of Developmental Biology, University of Colorado Denver School of Medicine, Aurora, CO, USA and
| | | |
Collapse
|
1050
|
The role of mesodermal signals during liver organogenesis in zebrafish. SCIENCE CHINA-LIFE SCIENCES 2010; 53:455-61. [PMID: 20596911 DOI: 10.1007/s11427-010-0078-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2009] [Accepted: 11/02/2009] [Indexed: 12/22/2022]
Abstract
Three germ cell layers, the ectoderm, mesoderm and endoderm, are established during the gastrulation stage. All cell types in different organs and tissues are derived from these 3 germ cell layers at later stages. For example, skin epithelial cells and neuronal cells are derived from the ectoderm, while endothelial cells and muscle cells from the mesoderm and lung, and intestine epithelial cells from the endoderm. While in a normal situation different germ cells are destined to specific cell fates in different organs and tissues, each type of germ cells or its derivatives also produce extracellular signaling molecules to direct and facilitate the specification and differentiation of other germ cells during organogenesis. Liver is derived from the endoderm, but completion of liver organogenesis is regulated at different levels. While the pan-endoderm factors (e.g. FoxA and Gata families) and liver specific factors (e.g. Prox1 and Hhex) are essential intrinsic factors for endoderm cells to be differentiated into hepatoblasts, the role of signals produced by neighboring mesoderm cells for liver organogenesis is equally important. This review summarizes recent progress in studying the role of Bone morphogenetic proteins (Bmp), Fibroblast growth factors (Fgf), retinoic acid (RA) and Wingless and Int (Wnt), the 4 types of signaling molecules produced by the mesoderm cells, in liver organogenesis in zebrafish.
Collapse
|