1
|
Gerges A, Canning U. High-Risk Neuroblastoma Stage 4 (NBS4): Developing a Medicinal Chemistry Multi-Target Drug Approach. Molecules 2025; 30:2211. [PMID: 40430383 PMCID: PMC12114419 DOI: 10.3390/molecules30102211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Childhood neuroblastoma (NB) is a malignant tumour that is a member of a class of embryonic tumours that have their origins in sympathoadrenal progenitor cells. There are five stages in the clinical NB staging system: 1, 2A, 2B, 3, 4S, and 4. For those diagnosed with stage 4 neuroblastoma (NBS4), the treatment options are limited with a survival rate of between 40 and 50%. Since 1975, more than 15 targets have been identified in the search for a treatment for high-risk NBS4. This article is concerned with the search for a multi-target drug treatment for high-risk NBS4 and focuses on four possible treatment targets that research has identified as having a role in the development of NBS4 and includes the inhibitors Histone Deacetylase (HDAC), Bromodomain (BRD), Hedgehog (HH), and Tropomyosin Kinase (TRK). Computer-aided drug design and molecular modelling have greatly assisted drug discovery in medicinal chemistry. Computational methods such as molecular docking, homology modelling, molecular dynamics, and quantitative structure-activity relationships (QSAR) are frequently used as part of the process for finding new therapeutic drug targets. Relying on these techniques, the authors describe a medicinal chemistry strategy that successfully identified eight compounds (inhibitors) that were thought to be potential inhibitors for each of the four targets listed above. Results revealed that all four targets BRD, HDAC, HH and TRK receptors binding sites share similar amino acid sequencing that ranges from 80 to 100%, offering the possibility of further testing for multi-target drug use. Two additional targets were also tested as part of this work, Retinoic Acid (RA) and c-Src (Csk), which showed similarity (of the binding pocket) across their receptors of 80-100% but lower than 80% for the other four targets. The work for these two targets is the subject of a paper currently in progress.
Collapse
Affiliation(s)
- Amgad Gerges
- School of Human Sciences, London Metropolitan University, 166-220 Holloway Rd., London N7 8DB, UK
| | | |
Collapse
|
2
|
Gerges A, Canning U. Neuroblastoma and its Target Therapies: A Medicinal Chemistry Review. ChemMedChem 2024; 19:e202300535. [PMID: 38340043 DOI: 10.1002/cmdc.202300535] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/22/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Neuroblastoma (NB) is a childhood malignant tumour belonging to a group of embryonic tumours originating from progenitor cells of the sympathoadrenal lineage. The heterogeneity of NB is reflected in the survival rates of those with low and intermediate risk diseases who have survival rates ranging from 85 to 90 %. However, for those identified with high-risk Stage 4 NB, the treatment options are much more limited. For this group, current treatment consists of immunotherapy (monoclonal antibodies) in combination with anti-cancer drugs and has a 40 to 50 % survival rate. The purpose of this review is to summarise NB research from a medicinal chemistry perspective and to highlight advances in targeted drug therapy in the field. The review examines the medicinal chemistry of a number of drugs tested in research, some of which are currently under clinical trial. It concludes by proposing that future medicinal chemistry research into NB should consider other possible target therapies and adopt a multi-target drug approach rather than a one-drug-one-target approach for improved efficacy and less drug-drug interaction for the treatment of NB Stage 4 (NBS4) patients.
Collapse
Affiliation(s)
- A Gerges
- Bioscience Department, London Metropolitan University, 166-220 Holloway Road, London, N7 8DB, England, United Kingdom
| | - U Canning
- Bioscience Department, London Metropolitan University, 166-220 Holloway Road, London, N7 8DB, England, United Kingdom
| |
Collapse
|
3
|
Sotcheff SL, Chen JYC, Elrod N, Cao J, Jaworski E, Kuyumcu-Martinez MN, Shi PY, Routh AL. Zika Virus Infection Alters Gene Expression and Poly-Adenylation Patterns in Placental Cells. Pathogens 2022; 11:pathogens11080936. [PMID: 36015056 PMCID: PMC9414685 DOI: 10.3390/pathogens11080936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/29/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Flaviviruses are small RNA viruses that are mainly transmitted via arthropod vectors and are found in tropic and sub-tropical regions. Most infections are asymptomatic (90-95%), but symptoms can be as severe as hemorrhagic fever and encephalitis. One recently emerged flavivirus is Zika virus (ZIKV), which was originally isolated from rhesus monkeys in Uganda roughly 70 years ago but has recently spread east, reaching S. America in 2015-2016. This outbreak was associated with the development of Guillain-Barré syndrome in adults and microcephaly in infants born to expectant mothers infected early in pregnancy. ZIKV must traverse the placenta to impact the development of the fetus, but the mechanisms responsible are unknown. While flaviviruses are known to disrupt splicing patterns in host cells, little is known about how flaviviruses such as ZIKV impact the alternative polyadenylation (APA) of host transcripts. This is important as APA is well-established as a mechanism in the regulation of mRNA metabolism and translation. Thus, we sought to characterize transcriptomic changes including APA in human placental (JEG3) cells in response to ZIKV infection using Poly(A)-ClickSeq (PAC-Seq). We used our differential Poly(A)-cluster (DPAC) analysis pipeline to characterize changes in differential gene expression, alternative poly-adenylation (APA) and the use of alternative terminal exons. We identified 98 upregulated genes and 28 downregulated genes. Pathway enrichment analysis indicated that many RNA processing and immune pathways were upregulated in ZIKV-infected JEG3 cells. We also updated DPAC to provide additional metrics of APA including the percentage-distal usage index (PDUI), which revealed that APA was extensive and the 3' UTRs of 229 genes were lengthened while 269 were shortened. We further found that there were 214 upregulated and 59 downregulated poly(A)-clusters (PACs). We extracted the nucleotide sequences surrounding these PACs and found that the canonical signals for poly-adenylation (binding site for poly-A binding protein (PABP) upstream and a GU-rich region down-stream of the PAC) were only enriched in the downregulated PACs. These results indicate that ZIKV infection makes JEG3 cells more permissive to non-canonical poly-adenylation signals.
Collapse
Affiliation(s)
- Stephanea L. Sotcheff
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - John Yun-Chung Chen
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nathan Elrod
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jun Cao
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Elizabeth Jaworski
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mugé N. Kuyumcu-Martinez
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Andrew L. Routh
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
- Correspondence:
| |
Collapse
|
4
|
Lai J, Liu Z, Zhao Y, Ma C, Huang H. Anticancer Effects of I-BET151, an Inhibitor of Bromodomain and Extra-Terminal Domain Proteins. Front Oncol 2021; 11:716830. [PMID: 34540687 PMCID: PMC8443787 DOI: 10.3389/fonc.2021.716830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/17/2021] [Indexed: 11/13/2022] Open
Abstract
I-BET151 is an inhibitor of bromodomain and extra-terminal domain (BET) proteins that selectively inhibits BET family members (BRD2, BRD3, BRD4, and BRDT). Over the past ten years, many studies have demonstrated the potential of I-BET151 in cancer treatment. Specifically, I-BET151 causes cell cycle arrest and inhibits tumor cell proliferation in some hematological malignancies and solid tumors, such as breast cancer, glioma, melanoma, neuroblastoma, and ovarian cancer. The anticancer activity of I-BET151 is related to its effects on NF-κB, Notch, and Hedgehog signal transduction pathway, tumor microenvironment (TME) and telomere elongation. Remarkably, the combination of I-BET151 with select anticancer drugs can partially alleviate the occurrence of drug resistance in chemotherapy. Especially, the combination of forskolin, ISX9, CHIR99021, I-BET151 and DAPT allows GBM cells to be reprogrammed into neurons, and this process does not experience an intermediate pluripotent state. The research on the anticancer mechanism of I-BET151 will lead to new treatment strategies for clinical cancer.
Collapse
Affiliation(s)
- Jiacheng Lai
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Ziqiang Liu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yulei Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Chengyuan Ma
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Haiyan Huang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Mechanism and ultrasensitivity in Hedgehog signaling revealed by Patched1 disease mutations. Proc Natl Acad Sci U S A 2021; 118:2006800118. [PMID: 33526656 DOI: 10.1073/pnas.2006800118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hedgehog signaling is fundamental in animal embryogenesis, and its dysregulation causes cancer and birth defects. The pathway is triggered when the Hedgehog ligand inhibits the Patched1 membrane receptor, relieving repression that Patched1 exerts on the GPCR-like protein Smoothened. While it is clear how loss-of-function Patched1 mutations cause hyperactive Hedgehog signaling and cancer, how other Patched1 mutations inhibit signaling remains unknown. Here, we develop quantitative single-cell functional assays for Patched1, which, together with mathematical modeling, indicate that Patched1 inhibits Smoothened enzymatically, operating in an ultrasensitive regime. Based on this analysis, we propose that Patched1 functions in cilia, catalyzing Smoothened deactivation by removing cholesterol bound to its extracellular, cysteine-rich domain. Patched1 mutants associated with holoprosencephaly dampen signaling by three mechanisms: reduced affinity for Hedgehog ligand, elevated catalytic activity, or elevated affinity for the Smoothened substrate. Our results clarify the enigmatic mechanism of Patched1 and explain how Patched1 mutations lead to birth defects.
Collapse
|
6
|
Curry RN, Glasgow SM. The Role of Neurodevelopmental Pathways in Brain Tumors. Front Cell Dev Biol 2021; 9:659055. [PMID: 34012965 PMCID: PMC8127784 DOI: 10.3389/fcell.2021.659055] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Disruptions to developmental cell signaling pathways and transcriptional cascades have been implicated in tumor initiation, maintenance and progression. Resurgence of aberrant neurodevelopmental programs in the context of brain tumors highlights the numerous parallels that exist between developmental and oncologic mechanisms. A deeper understanding of how dysregulated developmental factors contribute to brain tumor oncogenesis and disease progression will help to identify potential therapeutic targets for these malignancies. In this review, we summarize the current literature concerning developmental signaling cascades and neurodevelopmentally-regulated transcriptional programs. We also examine their respective contributions towards tumor initiation, maintenance, and progression in both pediatric and adult brain tumors and highlight relevant differentiation therapies and putative candidates for prospective treatments.
Collapse
Affiliation(s)
- Rachel N. Curry
- Department of Neuroscience, Baylor College of Medicine, Center for Cell and Gene Therapy, Houston, TX, United States
- Integrative Molecular and Biomedical Sciences, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX, United States
| | - Stacey M. Glasgow
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
- Neurosciences Graduate Program, University of California, San Diego, San Diego, CA, United States
- Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
7
|
Pan P. A Case of Split Notochord Syndrome with Left Congenital Diaphragmatic Hernia: A Rare Association. J Pediatr Neurosci 2020; 15:42-44. [PMID: 32435306 PMCID: PMC7227745 DOI: 10.4103/jpn.jpn_85_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/04/2019] [Accepted: 10/30/2019] [Indexed: 11/21/2022] Open
Abstract
Split notochord syndrome is a rare neural tube malformation involving the brain, spinal cord, and vertebral column. In nearly half of the cases, the malformation also involves the gastrointestinal tract in the form of a dorsal neurenteric fistula and imperforate anus. In the literature, less than 50 cases have been reported. To the best of my knowledge, this is the first reported case associated with a left congenital diaphragmatic hernia.
Collapse
Affiliation(s)
- Pradyumna Pan
- Pediatric Surgery Unit, Ashish Hospital and Research Centre, Jabalpur, Madhya Pradesh, India
| |
Collapse
|
8
|
Peer E, Tesanovic S, Aberger F. Next-Generation Hedgehog/GLI Pathway Inhibitors for Cancer Therapy. Cancers (Basel) 2019; 11:cancers11040538. [PMID: 30991683 PMCID: PMC6520835 DOI: 10.3390/cancers11040538] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/26/2022] Open
Abstract
The Hedgehog/Glioma-associated oncogene homolog (HH/GLI) signaling pathway regulates self-renewal of rare and highly malignant cancer stem cells (CSC), which have been shown to account for the initiation and maintenance of tumor growth as well as for drug resistance, metastatic spread and relapse. Efficacious therapeutic approaches targeting CSC pathways, such as HH/GLI signaling in combination with chemo, radiation or immunotherapy are, therefore, of high medical need. Pharmacological inhibition of HH/GLI pathway activity represents a promising approach to eliminate malignant CSC. Clinically approved HH/GLI pathway inhibitors target the essential pathway effector Smoothened (SMO) with striking therapeutic efficacy in skin and brain cancer patients. However, multiple genetic and molecular mechanisms resulting in de novo and acquired resistance to SMO inhibitors pose major limitations to anti-HH/GLI therapies and, thus, the eradication of CSC. In this review, we summarize reasons for clinical failure of SMO inhibitors, including mechanisms caused by genetic alterations in HH pathway effectors or triggered by additional oncogenic signals activating GLI transcription factors in a noncanonical manner. We then discuss emerging novel and rationale-based approaches to overcome SMO-inhibitor resistance, focusing on pharmacological perturbations of enzymatic modifiers of GLI activity and on compounds either directly targeting oncogenic GLI factors or interfering with synergistic crosstalk signals known to boost the oncogenicity of HH/GLI signaling.
Collapse
Affiliation(s)
- Elisabeth Peer
- Department of Biosciences, Paris-Lodron University of Salzburg, Cancer Cluster Salzburg, Hellbrunner Strasse 34, 5020 Salzburg, Austria.
| | - Suzana Tesanovic
- Department of Biosciences, Paris-Lodron University of Salzburg, Cancer Cluster Salzburg, Hellbrunner Strasse 34, 5020 Salzburg, Austria.
| | - Fritz Aberger
- Department of Biosciences, Paris-Lodron University of Salzburg, Cancer Cluster Salzburg, Hellbrunner Strasse 34, 5020 Salzburg, Austria.
| |
Collapse
|
9
|
Salaritabar A, Berindan-Neagoe I, Darvish B, Hadjiakhoondi F, Manayi A, Devi KP, Barreca D, Orhan IE, Süntar I, Farooqi AA, Gulei D, Nabavi SF, Sureda A, Daglia M, Dehpour AR, Nabavi SM, Shirooie S. Targeting Hedgehog signaling pathway: Paving the road for cancer therapy. Pharmacol Res 2019; 141:466-480. [DOI: 10.1016/j.phrs.2019.01.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/24/2018] [Accepted: 01/08/2019] [Indexed: 02/08/2023]
|
10
|
Allahyari RV, Clark KL, Shepard KA, Garcia ADR. Sonic hedgehog signaling is negatively regulated in reactive astrocytes after forebrain stab injury. Sci Rep 2019; 9:565. [PMID: 30679745 PMCID: PMC6345977 DOI: 10.1038/s41598-018-37555-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 12/07/2018] [Indexed: 12/19/2022] Open
Abstract
Following injury to the central nervous system, astrocytes perform critical and complex functions that both promote and antagonize neural repair. Understanding the molecular signaling pathways that coordinate their diverse functional properties is key to developing effective therapeutic strategies. In the healthy, adult CNS, Sonic hedgehog (Shh) signaling is active in mature, differentiated astrocytes. Shh has been shown to undergo injury-induced upregulation and promote neural repair. Here, we investigated whether Shh signaling mediates astrocyte response to injury. Surprisingly, we found that following an acute, focal injury, reactive astrocytes exhibit a pronounced reduction in Shh activity in a spatiotemporally-defined manner. Shh signaling is lost in reactive astrocytes at the lesion site, but persists in mild to moderately reactive astrocytes in distal tissues. Nevertheless, local pharmacological activation of the Shh pathway in astrocytes mitigates inflammation, consistent with a neuroprotective role for Shh signaling after injury. Interestingly, we find that Shh signaling is restored to baseline levels two weeks after injury, a time during which acute inflammation has largely subsided and lesions have matured. Taken together, these data suggest that endogenous Shh signaling in astrocytes is dynamically regulated in a context dependent manner. In addition, exogenous activation of the Shh pathway promotes neuroprotection mediated by reactive astrocytes.
Collapse
Affiliation(s)
- R Vivian Allahyari
- Departments of Biology and Neurobiology and Anatomy, Drexel University, Philadelphia, PA, 19104, USA
| | - K Lyles Clark
- Departments of Biology and Neurobiology and Anatomy, Drexel University, Philadelphia, PA, 19104, USA
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Katherine A Shepard
- Departments of Biology and Neurobiology and Anatomy, Drexel University, Philadelphia, PA, 19104, USA
| | - A Denise R Garcia
- Departments of Biology and Neurobiology and Anatomy, Drexel University, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Pindiprolu SKSS, Krishnamurthy PT, Chintamaneni PK. Pharmacological targets of breast cancer stem cells: a review. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:463-479. [PMID: 29476201 DOI: 10.1007/s00210-018-1479-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 02/13/2018] [Indexed: 02/07/2023]
Abstract
Breast cancers contain small population of tumor-initiating cells called breast cancer stem cells (BCSCs), which are spared even after chemotherapy. Recently, BCSCs are implicated to be a cause of metastasis, tumor relapse, and therapy resistance in breast cancer. BCSCs have unique molecular mechanisms, which can be targeted to eliminate them. These include surface biomarkers, proteins involved in self-renewal pathways, drug efflux transporters, apoptotic/antiapoptotic proteins, autophagy, metabolism, and microenvironment regulation. The complex molecular mechanisms behind the survival of BCSCs and pharmacological targets for elimination of BCSCs are described in this review.
Collapse
Affiliation(s)
- Sai Kiran S S Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India.
| | - Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| |
Collapse
|
12
|
Driver AM, Shumrick C, Stottmann RW. Ttc21b Is Required in Bergmann Glia for Proper Granule Cell Radial Migration. J Dev Biol 2017; 5:E18. [PMID: 29615573 PMCID: PMC5831799 DOI: 10.3390/jdb5040018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 11/30/2017] [Accepted: 12/11/2017] [Indexed: 12/11/2022] Open
Abstract
Proper cerebellar development is dependent on tightly regulated proliferation, migration, and differentiation events. Disruptions in any of these leads to a range of cerebellar phenotypes from ataxia to childhood tumors. Animal models have shown that proper regulation of sonic hedgehog (Shh) signaling is crucial for normal cerebellar architecture, and increased signaling leads to cerebellar tumor formation. Primary cilia are known to be required for the proper regulation of multiple developmental signaling pathways, including Shh. Tetratricopeptide Repeat Domain 21B (Ttc21b) is required for proper primary cilia form and function, and is primarily thought to restrict Shh signaling. Here we investigated a role for Ttc21b in cerebellar development. Surprisingly, Ttc21b ablation in Bergmann glia resulted in the accumulation of ectopic granule cells in the lower/posterior lobes of the cerebellum and a reduction in Shh signaling. Ttc21b ablation in just Purkinje cells resulted in a similar phenotype seen in fewer cells, but across the entire extent of the cerebellum. These results suggest that Ttc21b expression is required for Bergmann glia structure and signaling in the developing cerebellum, and in some contexts, augments rather than attenuates Shh signaling.
Collapse
Affiliation(s)
- Ashley M Driver
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Christopher Shumrick
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Rolf W Stottmann
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
13
|
Bhat KM. Post-guidance signaling by extracellular matrix-associated Slit/Slit-N maintains fasciculation and position of axon tracts in the nerve cord. PLoS Genet 2017; 13:e1007094. [PMID: 29155813 PMCID: PMC5714384 DOI: 10.1371/journal.pgen.1007094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 12/04/2017] [Accepted: 11/01/2017] [Indexed: 11/19/2022] Open
Abstract
Axon-guidance by Slit-Roundabout (Robo) signaling at the midline initially guides growth cones to synaptic targets and positions longitudinal axon tracts in discrete bundles on either side of the midline. Following the formation of commissural tracts, Slit is found also in tracts of the commissures and longitudinal connectives, the purpose of which is not clear. The Slit protein is processed into a larger N-terminal peptide and a smaller C-terminal peptide. Here, I show that Slit and Slit-N in tracts interact with Robo to maintain the fasciculation, the inter-tract spacing between tracts and their position relative to the midline. Thus, in the absence of Slit in post-guidance tracts, tracts de-fasciculate, merge with one another and shift their position towards the midline. The Slit protein is proposed to function as a gradient. However, I show that Slit and Slit-N are not freely present in the extracellular milieu but associated with the extracellular matrix (ECM) and both interact with Robo1. Slit-C is tightly associated with the ECM requiring collagenase treatment to release it, and it does not interact with Robo1. These results define a role for Slit and Slit-N in tracts for the maintenance and fasciculation of tracts, thus the maintenance of the hardwiring of the CNS.
Collapse
Affiliation(s)
- Krishna Moorthi Bhat
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch School of Medicine, Galveston, Texas, United States of America
| |
Collapse
|
14
|
Abstract
Communication between cells pervades the development and physiology of metazoans. In animals, this process is carried out by a relatively small number of signaling pathways, each consisting of a chain of biochemical events through which extracellular stimuli control the behavior of target cells. One such signaling system is the Hedgehog pathway, which is crucial in embryogenesis and is implicated in many birth defects and cancers. Although Hedgehog pathway components were identified by genetic analysis more than a decade ago, our understanding of the molecular mechanisms of signaling is far from complete. In this review, we focus on the biochemistry and cell biology of the Hedgehog pathway. We examine the unique biosynthesis of the Hedgehog ligand, its specialized release from cells into extracellular space, and the poorly understood mechanisms involved in ligand reception and pathway activation at the surface of target cells. We highlight several critical questions that remain open.
Collapse
Affiliation(s)
- Kostadin Petrov
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115; ,
| | - Bradley M Wierbowski
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115; ,
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115; ,
| |
Collapse
|
15
|
Wessler S, Krisch LM, Elmer DP, Aberger F. From inflammation to gastric cancer - the importance of Hedgehog/GLI signaling in Helicobacter pylori-induced chronic inflammatory and neoplastic diseases. Cell Commun Signal 2017; 15:15. [PMID: 28427431 PMCID: PMC5397778 DOI: 10.1186/s12964-017-0171-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/07/2017] [Indexed: 02/07/2023] Open
Abstract
Infections with the human pathogen Helicobacter pylori (H. pylori) are closely associated with the development of inflammatory disorders and neoplastic transformation of the gastric epithelium. Drastic changes in the micromilieu involve a complex network of H. pylori-regulated signal transduction pathways leading to the release of proinflammatory cytokines, gut hormones and a wide range of signaling molecules. Besides controlling embryonic development, the Hedgehog/GLI signaling pathway also plays important roles in epithelial proliferation, differentiation, and regeneration of the gastric physiology, but also in the induction and progression of inflammation and neoplastic transformation in H. pylori infections. Here, we summarize recent findings of H. pylori-associated Hedgehog/GLI signaling in gastric homeostasis, malignant development and the modulation of the gastric tumor microenvironment.
Collapse
Affiliation(s)
- Silja Wessler
- Division of Microbiology, Cancer Cluster Salzburg, Department of Molecular Biology, Paris-Lodron University of Salzburg, Billroth Strasse 11, A-5020, Salzburg, Austria.
| | - Linda M Krisch
- Division of Microbiology, Cancer Cluster Salzburg, Department of Molecular Biology, Paris-Lodron University of Salzburg, Billroth Strasse 11, A-5020, Salzburg, Austria
| | - Dominik P Elmer
- Division of Molecular Tumor Biology, Cancer Cluster Salzburg, Department of Molecular Biology, Paris-Lodron University of Salzburg, Hellbrunner Strasse 34, A-5020, Salzburg, Austria
| | - Fritz Aberger
- Division of Molecular Tumor Biology, Cancer Cluster Salzburg, Department of Molecular Biology, Paris-Lodron University of Salzburg, Hellbrunner Strasse 34, A-5020, Salzburg, Austria.
| |
Collapse
|
16
|
Gueta K, David A, Cohen T, Menuchin-Lasowski Y, Nobel H, Narkis G, Li L, Love P, de Melo J, Blackshaw S, Westphal H, Ashery-Padan R. The stage-dependent roles of Ldb1 and functional redundancy with Ldb2 in mammalian retinogenesis. Development 2016; 143:4182-4192. [PMID: 27697904 DOI: 10.1242/dev.129734] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/20/2016] [Indexed: 12/26/2022]
Abstract
The Lim domain-binding proteins are key co-factor proteins that assemble with LIM domains of the LMO/LIM-HD family to form functional complexes that regulate cell proliferation and differentiation. Using conditional mutagenesis and comparative phenotypic analysis, we analyze the function of Ldb1 and Ldb2 in mouse retinal development, and demonstrate overlapping and specific functions of both proteins. Ldb1 interacts with Lhx2 in the embryonic retina and both Ldb1 and Ldb2 play a key role in maintaining the pool of retinal progenitor cells. This is accomplished by controlling the expression of the Vsx2 and Rax, and components of the Notch and Hedgehog signaling pathways. Furthermore, the Ldb1/Ldb2-mediated complex is essential for generation of early-born photoreceptors through the regulation of Rax and Crx. Finally, we demonstrate functional redundancy between Ldb1 and Ldb2. Ldb1 can fully compensate the loss of Ldb2 during all phases of retinal development, whereas Ldb2 alone is sufficient to sustain activity of Lhx2 in both early- and late-stage RPCs and in Müller glia. By contrast, loss of Ldb1 disrupts activity of the LIM domain factors in neuronal precursors. An intricate regulatory network exists that is mediated by Ldb1 and Ldb2, and promotes RPC proliferation and multipotency; it also controls specification of mammalian retina cells.
Collapse
Affiliation(s)
- Keren Gueta
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ahuvit David
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tsadok Cohen
- Mammalian Genes and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yotam Menuchin-Lasowski
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Hila Nobel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ginat Narkis
- Mammalian Genes and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - LiQi Li
- Program on Genomics of Differentiation, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul Love
- Program on Genomics of Differentiation, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jimmy de Melo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Heiner Westphal
- Mammalian Genes and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
17
|
SERPINE2/Protease Nexin-1 in vivo multiple functions: Does the puzzle make sense? Semin Cell Dev Biol 2016; 62:160-169. [PMID: 27545616 DOI: 10.1016/j.semcdb.2016.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/17/2016] [Accepted: 08/17/2016] [Indexed: 11/21/2022]
Abstract
Cultures of glial cells and fibroblasts allowed and lead to the identification SERPINE2/Protease Nexin-1 (SERPINE2/PN-1). Cellular, biochemical, immunological and molecular characterization substantiated its variable expression in many organs as a function of development, adult stages, pathological situations or following injury. It is not a circulating serpin, but as other members of the family, its target specificity is influenced by components of the extracellular matrix. The challenges are to identify where and when SERPINE2/PN-1 modulatory action becomes crucial or even possibly specific in a mosaic of feasible in vivo impacts. Data providing correlations are not sufficient to satisfy this aim. Genetically modified mice, or tissue derived thereof, provide interesting in vivo models to identify and study the relevance of this serpin. This review will highlight sometimes-intriguing results indicating a crucial impact of SERPINE2/PN-1, especially in the vasculature, the nervous system or the behavior of cancer cells in vivo. Data presently available will be discussed in an attempt to define general trends in the diversity of SERPINE2/PN-1 modes of action in vivo.
Collapse
|
18
|
Klein C, Zwick A, Kissel S, Forster CU, Pfeifer D, Follo M, Illert AL, Decker S, Benkler T, Pahl H, Oostendorp RAJ, Aumann K, Duyster J, Dierks C. Ptch2 loss drives myeloproliferation and myeloproliferative neoplasm progression. J Exp Med 2016; 213:273-90. [PMID: 26834157 PMCID: PMC4749921 DOI: 10.1084/jem.20150556] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 12/21/2015] [Indexed: 01/12/2023] Open
Abstract
Klein et al. show that Ptch2 loss in either the niche or in hematopoietic cells drives myeloproliferation and accelerates JAK2V617F-driven pathogenesis, causing transformation of nonlethal chronic MPNs into aggressive lethal leukemias. JAK2V617F+ myeloproliferative neoplasms (MPNs) frequently progress into leukemias, but the factors driving this process are not understood. Here, we find excess Hedgehog (HH) ligand secretion and loss of PTCH2 in myeloproliferative disease, which drives canonical and noncanonical HH-signaling. Interestingly, Ptch2−/− mice mimic dual pathway activation and develop a MPN-phenotype with leukocytosis (neutrophils and monocytes), strong progenitor and LKS mobilization, splenomegaly, anemia, and loss of lymphoid lineages. HSCs exhibit increased cell cycling with improved stress hematopoiesis after 5-FU treatment, and this results in HSC exhaustion over time. Cytopenias, LKS loss, and mobilization are all caused by loss of Ptch2 in the niche, whereas hematopoietic loss of Ptch2 drives leukocytosis and promotes LKS maintenance and replating capacity in vitro. Ptch2−/− niche cells show hyperactive noncanonical HH signaling, resulting in reduced production of essential HSC regulators (Scf, Cxcl12, and Jag1) and depletion of osteoblasts. Interestingly, Ptch2 loss in either the niche or in hematopoietic cells dramatically accelerated human JAK2V617F-driven pathogenesis, causing transformation of nonlethal chronic MPNs into aggressive lethal leukemias with >30% blasts in the peripheral blood. Our findings suggest HH ligand inhibitors as possible drug candidates that act on hematopoiesis and the niche to prevent transformation of MPNs into leukemias.
Collapse
Affiliation(s)
- Claudius Klein
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany Institute for Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - Anabel Zwick
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Sandra Kissel
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Christine Ulrike Forster
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Marie Follo
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Anna Lena Illert
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Sarah Decker
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Thomas Benkler
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Heike Pahl
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Robert A J Oostendorp
- Klinikum rechts der Isar der Technischen Universität München, III. Medizinische Klinik, 81675 München, Germany
| | - Konrad Aumann
- Department of Pathology, University Medical Center Freiburg, D-79104 Freiburg, Germany
| | - Justus Duyster
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Christine Dierks
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| |
Collapse
|
19
|
De Luca A, Cerrato V, Fucà E, Parmigiani E, Buffo A, Leto K. Sonic hedgehog patterning during cerebellar development. Cell Mol Life Sci 2016; 73:291-303. [PMID: 26499980 PMCID: PMC11108499 DOI: 10.1007/s00018-015-2065-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/06/2015] [Accepted: 10/08/2015] [Indexed: 01/30/2023]
Abstract
The morphogenic factor sonic hedgehog (Shh) actively orchestrates many aspects of cerebellar development and maturation. During embryogenesis, Shh signaling is active in the ventricular germinal zone (VZ) and represents an essential signal for proliferation of VZ-derived progenitors. Later, Shh secreted by Purkinje cells sustains the amplification of postnatal neurogenic niches: the external granular layer and the prospective white matter, where excitatory granule cells and inhibitory interneurons are produced, respectively. Moreover, Shh signaling affects Bergmann glial differentiation and promotes cerebellar foliation during development. Here we review the most relevant functions of Shh during cerebellar ontogenesis, underlying its role in physiological and pathological conditions.
Collapse
Affiliation(s)
- Annarita De Luca
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Elisa Fucà
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Elena Parmigiani
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Ketty Leto
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy.
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043, Orbassano, Turin, Italy.
| |
Collapse
|
20
|
Borah A, Raveendran S, Rochani A, Maekawa T, Kumar DS. Targeting self-renewal pathways in cancer stem cells: clinical implications for cancer therapy. Oncogenesis 2015; 4:e177. [PMID: 26619402 PMCID: PMC4670961 DOI: 10.1038/oncsis.2015.35] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/10/2015] [Accepted: 09/22/2015] [Indexed: 12/21/2022] Open
Abstract
Extensive cancer research in the past few decades has identified the existence of a rare subpopulation of stem cells in the grove of cancer cells. These cells are known as the cancer stem cells marked by the presence of surface biomarkers, multi-drug resistance pumps and deregulated self-renewal pathways (SRPs). They have a crucial role in provoking cancer cells leading to tumorigenesis and its progressive metastasis. Cancer stem cells (CSCs) are much alike to normal stem cells in their self-renewal mechanisms. However, deregulations in the SRPs are seen in CSCs, making them resistant to conventional chemotherapeutic agents resulting in the tumor recurrence. Current treatment strategies in cancer fail to detect and differentiate the CSCs from their non-tumorigenic progenies owing to absence of specific biomarkers. Now, it has become imperative to understand complex functional biology of CSCs, especially the signaling pathways to design improved treatment strategies to target them. It is hopeful that the SRPs in CSCs offer a promising target to alter their survival strategies and impede their tumorigenic potential. However, there are many perils associated with the direct targeting method by conventional therapeutic agents such as off targets, poor bioavailability and poor cellular distribution. Recent evidences have shown an increased use of small molecule antagonists directly to target these SRPs may lead to severe side-effects. An alternative to solve these issues could be an appropriate nanoformulation. Nanoformulations of these molecules could provide an added advantage for the selective targeting of the pathways especially Hedgehog, Wnt, Notch and B-cell-specific moloney murine leukemia virus integration site 1 in the CSCs while sparing the normal stem cells. Hence, to achieve this goal a complete understanding of the molecular pathways corroborate with the use of holistic nanosystem (nanomaterial inhibition molecule) could possibly be an encouraging direction for future cancer therapy.
Collapse
Affiliation(s)
- A Borah
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - S Raveendran
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - A Rochani
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - T Maekawa
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - D S Kumar
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| |
Collapse
|
21
|
Hou L, Dong Q, Wu YJ, Sun YX, Guo YY, Huo YH. Gonadotropins facilitate potential differentiation of human bone marrow mesenchymal stem cells into Leydig cells in vitro. Kaohsiung J Med Sci 2015; 32:1-9. [PMID: 26853168 DOI: 10.1016/j.kjms.2015.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/02/2015] [Accepted: 09/16/2015] [Indexed: 02/05/2023] Open
Abstract
Infertility due to low testosterone levels has increased in recent years. This has impacted the social well-being of the patients. This study was undertaken to investigate the potential of gonadotropins in facilitating differentiation of human bone marrow mesenchymal stem cells (BMSCs) into Leydig cells in vitro. BMSCs were isolated, cultured, and their biological characteristics were observed. BMSCs were induced with gonadotropins in vitro and their ability to differentiate into Leydig cells was studied. The level of expression of 3-beta hydroxysteroid dehydrogenase (3β-HSD) and secretion of testosterone were determined using flow cytometry and enzyme-linked immunosorbent assay, respectively, and the results were compared between the experimental and control groups. The cultured BMSCs showed a typical morphology of the fibroblast-like colony. The growth curve of cells formed an S-shape. After inducing the cells for 8-13 days, the cells in the experimental group increased in size and showed typical characteristics of Leydig cells, and the growth occurred in spindle or stellate shapes. Cells from the experimental group highly expressed 3β-HSD, and there was a gradual increase in the number of Leydig cells. The control group did not express 3β-HSD. The level of testosterone in the experimental group was higher than the control group (p < 0.05). Additionally, the cells in the experimental group secreted higher levels of testosterone with increased culture time. The expression of Leydig cell-specific markers in the experimental group was significantly higher (p < 0.05). With these findings, BMSCs can be considered a new approach for the treatment of patients with low androgen levels.
Collapse
Affiliation(s)
- Lin Hou
- Department of Urology, Number 5 Hospital of Datong, Datong, China
| | - Qiang Dong
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China.
| | - Yun-Jian Wu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan-Xing Sun
- Department of Urology, Number 5 Hospital of Datong, Datong, China
| | - Yan-Yu Guo
- Department of Rheumatology, Number 5 Hospital of Datong, Datong, China
| | - Yue-Hong Huo
- Department of Rheumatology, Number 5 Hospital of Datong, Datong, China
| |
Collapse
|
22
|
Callahan BP, Wang C. Hedgehog Cholesterolysis: Specialized Gatekeeper to Oncogenic Signaling. Cancers (Basel) 2015; 7:2037-53. [PMID: 26473928 PMCID: PMC4695875 DOI: 10.3390/cancers7040875] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/22/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022] Open
Abstract
Discussions of therapeutic suppression of hedgehog (Hh) signaling almost exclusively focus on receptor antagonism; however, hedgehog's biosynthesis represents a unique and potentially targetable aspect of this oncogenic signaling pathway. Here, we review a key biosynthetic step called cholesterolysis from the perspectives of structure/function and small molecule inhibition. Cholesterolysis, also called cholesteroylation, generates cholesterol-modified Hh ligand via autoprocessing of a hedgehog precursor protein. Post-translational modification by cholesterol appears to be restricted to proteins in the hedgehog family. The transformation is essential for Hh biological activity and upstream of signaling events. Despite its decisive role in generating ligand, cholesterolysis remains conspicuously unexplored as a therapeutic target.
Collapse
Affiliation(s)
- Brian P Callahan
- Chemistry Department, Binghamton University 4400 Vestal Parkway East, Binghamton, NY 13902, USA.
| | - Chunyu Wang
- Biology Department, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA.
| |
Collapse
|
23
|
Hartenstein V, Reh TA. Homologies between vertebrate and invertebrate eyes. Results Probl Cell Differ 2015; 37:219-55. [PMID: 25707078 DOI: 10.1007/978-3-540-45398-7_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Affiliation(s)
- Volker Hartenstein
- Department of Biology, University of California, Los Angeles, California, USA
| | | |
Collapse
|
24
|
Choe JY, Yun JY, Jeon YK, Kim SH, Choung HK, Oh S, Park M, Kim JE. Sonic hedgehog signalling proteins are frequently expressed in retinoblastoma and are associated with aggressive clinicopathological features. J Clin Pathol 2014; 68:6-11. [PMID: 25296932 DOI: 10.1136/jclinpath-2014-202434] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS This study aimed to examine the expression of Sonic hedgehog (SHH) signalling proteins in retinoblastoma and to evaluate its clinical significance. METHODS Seventy-nine enucleated retinoblastoma tumours were investigated immunohistochemically using antibodies against SHH pathway proteins, such as SHH, glioma-associated oncogene homologue (GLI) 1, GLI2, GLI3 and ABC binding cassette G2 (ABCG2). Western blotting of SHH signalling proteins was performed in two retinoblastoma cell lines. RESULTS SHH was expressed in most retinoblastoma cases (78 of 79, 98.7%), with 21 cases (26.6%) showing strong expression. GLI1 and GLI2 were also frequently expressed: 67 of 78 cases (85.9%) and 71 of 77 cases (92.2%), respectively. GLI3, a transcriptional repressor, was expressed at low levels in 23 of the 78 cases (29.5%). High ABCG2 expression was found in 23 of the 78 cases (29.5%). High expression levels of these proteins in retinoblastoma cell lines were confirmed by western blotting. The expression of SHH was associated with advanced stages, local invasion and metastasis (all p<0.05). CONCLUSIONS SHH signalling molecules were frequently expressed in retinoblastoma tumour cells, and high SHH expression was closely related to an advanced disease status. Our results suggest that the SHH signalling pathway may play a role in the progression of retinoblastoma.
Collapse
Affiliation(s)
- Ji-Young Choe
- Department of Pathology, Seoul National University, College of Medicine, Seoul, Korea Department of Pathology, Seoul National University Bundang Hospital, Gyeonggi-Do, Korea
| | - Ji Yun Yun
- Department of Pathology, Seoul National University, College of Medicine, Seoul, Korea Department of Pathology, Seoul National University Bundang Hospital, Gyeonggi-Do, Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University, College of Medicine, Seoul, Korea Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei Unversity, College of Medicine, Seoul, Korea
| | - Ho-Kyung Choung
- Department of Ophthalmology, Seoul National University Boramae Hospital, Seoul, Korea
| | - Sohee Oh
- Department of Biostatistics, Seoul National University Boramae Hospital, Seoul, Korea
| | - Mira Park
- Department of Pathology, Seoul National University Boramae Hospital, Seoul, Korea
| | - Ji Eun Kim
- Department of Pathology, Seoul National University, College of Medicine, Seoul, Korea Department of Pathology, Seoul National University Boramae Hospital, Seoul, Korea
| |
Collapse
|
25
|
Singh S, Chellappan S. Lung cancer stem cells: Molecular features and therapeutic targets. Mol Aspects Med 2014; 39:50-60. [PMID: 24016594 PMCID: PMC3949216 DOI: 10.1016/j.mam.2013.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 08/29/2013] [Accepted: 08/29/2013] [Indexed: 12/28/2022]
Abstract
Lung cancers are highly heterogeneous and resistant to available therapeutic agents, with a five year survival rate of less than 15%. Despite significant advances in our knowledge of the genetic alterations and aberrations in signaling pathways, it has been difficult to determine the basis of lung cancer heterogeneity and drug resistance. Cancer stem cell model has attracted a significant amount of attention in recent years as a viable explanation for the heterogeneity, drug resistance, dormancy and recurrence and metastasis of various tumors. At the same time, cancer stem cells have been relatively less characterized in lung cancers. This review summarizes the current understanding of lung cancer stem cells, including their molecular features and signaling pathways that drive their stemness. This review also discusses the potential startegies to inhibit the signaling pathways driving stemness, in an effort to eradicate these cells to combat lung cancer.
Collapse
Affiliation(s)
- Sandeep Singh
- National Institute of Biomedical Genomics (NIBMG), TB Hospital Building, 2nd floor, Kalyani, West Bengal, India
| | - Srikumar Chellappan
- Department of Tumor Biology, H. Lee Moffitt cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, United States.
| |
Collapse
|
26
|
Jenkins NC, Kalra RR, Dubuc A, Sivakumar W, Pedone CA, Wu X, Taylor MD, Fults DW. Genetic drivers of metastatic dissemination in sonic hedgehog medulloblastoma. Acta Neuropathol Commun 2014; 2:85. [PMID: 25059231 PMCID: PMC4149244 DOI: 10.1186/s40478-014-0085-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 01/31/2023] Open
Abstract
Leptomeningeal dissemination (LMD), the metastatic spread of tumor cells via the cerebrospinal fluid to the brain and spinal cord, is an ominous prognostic sign for patients with the pediatric brain tumor medulloblastoma. The need to reduce the risk of LMD has driven the development of aggressive treatment regimens, which cause disabling neurotoxic side effects in long-term survivors. Transposon-mediated mutagenesis studies in mice have revealed numerous candidate metastasis genes. Understanding how these genes drive LMD will require functional assessment using in vivo and cell culture models of medulloblastoma. We analyzed two genes that were sites of frequent transposon insertion and highly expressed in human medulloblastomas: Arnt (aryl hydrocarbon receptor nuclear translocator) and Gdi2 (GDP dissociation inhibitor 2). Here we show that ectopic expression of Arnt and Gdi2 promoted LMD in mice bearing Sonic hedgehog (Shh)-induced medulloblastomas. We overexpressed Arnt and Gdi2 in a human medulloblastoma cell line (DAOY) and an immortalized, nontransformed cell line derived from mouse granule neuron precursors (SHH-NPD) and quantified migration, invasiveness, and anchorage-independent growth, cell traits that are associated with metastatic competence in carcinomas. In SHH-NPD cells. Arnt and Gdi2 stimulated all three traits. In DAOY cells, Arnt had the same effects, but Gdi2 stimulated invasiveness only. These results support a mechanism whereby Arnt and Gdi2 cause cells to detach from the primary tumor mass by increasing cell motility and invasiveness. By conferring to tumor cells the ability to proliferate without surface attachment, Arnt and Gdi2 favor the formation of stable colonies of cells capable of seeding the leptomeninges.
Collapse
|
27
|
Cerri F, Salvatore L, Memon D, Boneschi FM, Madaghiele M, Brambilla P, Del Carro U, Taveggia C, Riva N, Trimarco A, Lopez ID, Comi G, Pluchino S, Martino G, Sannino A, Quattrini A. Peripheral nerve morphogenesis induced by scaffold micropatterning. Biomaterials 2014; 35:4035-4045. [PMID: 24559639 DOI: 10.1016/j.biomaterials.2014.01.069] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/26/2014] [Indexed: 10/25/2022]
Abstract
Several bioengineering approaches have been proposed for peripheral nervous system repair, with limited results and still open questions about the underlying molecular mechanisms. We assessed the biological processes that occur after the implantation of collagen scaffold with a peculiar porous micro-structure of the wall in a rat sciatic nerve transection model compared to commercial collagen conduits and nerve crush injury using functional, histological and genome wide analyses. We demonstrated that within 60 days, our conduit had been completely substituted by a normal nerve. Gene expression analysis documented a precise sequential regulation of known genes involved in angiogenesis, Schwann cells/axons interactions and myelination, together with a selective modulation of key biological pathways for nerve morphogenesis induced by porous matrices. These data suggest that the scaffold's micro-structure profoundly influences cell behaviors and creates an instructive micro-environment to enhance nerve morphogenesis that can be exploited to improve recovery and understand the molecular differences between repair and regeneration.
Collapse
Affiliation(s)
- Federica Cerri
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Luca Salvatore
- Department of Innovation Engineering, University of Lecce, Via per Monteroni, 73100 Lecce, Italy
| | - Danish Memon
- Department of Clinical Neurosciences, Centre for Brain Repair, University of Cambridge, Robinson Way CB2 0PY, UK
| | | | - Marta Madaghiele
- Department of Innovation Engineering, University of Lecce, Via per Monteroni, 73100 Lecce, Italy
| | - Paola Brambilla
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Ubaldo Del Carro
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Carla Taveggia
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Nilo Riva
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Amelia Trimarco
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Ignazio D Lopez
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Giancarlo Comi
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Stefano Pluchino
- Department of Clinical Neurosciences, Centre for Brain Repair, University of Cambridge, Robinson Way CB2 0PY, UK
| | - Gianvito Martino
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Alessandro Sannino
- Department of Innovation Engineering, University of Lecce, Via per Monteroni, 73100 Lecce, Italy
| | - Angelo Quattrini
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| |
Collapse
|
28
|
Henner A, Ventura PB, Jiang Y, Zong H. MADM-ML, a mouse genetic mosaic system with increased clonal efficiency. PLoS One 2013; 8:e77672. [PMID: 24143253 PMCID: PMC3797059 DOI: 10.1371/journal.pone.0077672] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 09/11/2013] [Indexed: 01/07/2023] Open
Abstract
Mosaic Analysis with Double Markers (MADM) is a mouse genetic system that allows simultaneous gene knockout and fluorescent labeling of sparse, clonally-related cells within an otherwise normal mouse, thereby circumventing embryonic lethality problems and providing single-cell resolution for phenotypic analysis in vivo. The clonal efficiency of MADM is intrinsically low because it relies on Cre/loxP-mediated mitotic recombination between two homologous chromosomes rather than within the same chromosome, as in the case of conditional knockout (CKO). Although sparse labeling enhances in vivo resolution, the original MADM labels too few or even no cells when a low-expressing Cre transgene is used or a small population of cells is studied. Recently, we described the usage of a new system, MADM-ML, which contains three mutually exclusive, self-recognizing loxP variant sites as opposed to a single loxP site present in the original MADM system (referred to as MADM-SL in this paper). Here we carefully compared the recombination efficiency between MADM-SL and MADM-ML using the same Cre transgene, and found that the new system labels significantly more cells than the original system does. When we established mouse medulloblastoma models with both the original and the new MADM systems, we found that, while the MADM-SL model suffered from varied tumor progression and incomplete penetrance, the MADM-ML model had consistent tumor progression and full penetrance of tumor formation. Therefore MADM-ML, with its higher recombination efficiency, will broaden the applicability of MADM for studying many biological questions including normal development and disease modeling at cellular resolution in vivo.
Collapse
Affiliation(s)
- Astra Henner
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
| | - P. Britten Ventura
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Ying Jiang
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Hui Zong
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
- * E-mail:
| |
Collapse
|
29
|
de Weille J, Fabre C, Bakalara N. Oxysterols in cancer cell proliferation and death. Biochem Pharmacol 2013; 86:154-60. [PMID: 23500545 DOI: 10.1016/j.bcp.2013.02.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 02/27/2013] [Accepted: 02/27/2013] [Indexed: 12/21/2022]
Abstract
Oxysterols have been shown to interfere with proliferation and cause the death of many cancer cell types, such as leukaemia, glioblastoma, colon, breast and prostate cancer cells, while they have little or no effect on senescent cells. The mechanisms by which oxysterols may influence proliferation are manifold: they control the transcription and the turnover of the key enzyme in cholesterol synthesis, 3-hydroxy-3-methylglutaryl CoA reductase, by binding to Insig-1, Insig-2 and liver X receptors. Oxysterols are thought to be generated in proportion to the rate of cholesterol synthesis. Although there is no consensus about the mechanism by which these oxysterols are generated in vivo, it clearly has to be ubiquitous. The 25- and the 27-cholesterol hydroxylases, present in almost all tissues, are possible candidates. Cholesterol uptake from lipoproteins, intracellular vesicle transport and lipid transfer are also modified by oxysterols. Oxysterols interfere with ERK, hedgehog and wnt pathways of proliferation and differentiation. When administered in vitro to cancer cell lines, oxysterols invariably both slow down proliferation and provoke cell death. Perhaps is it sufficient to stop proliferation of a cancer to provoke its eradication. Therefore, the two facets of oxysterol action that seem important for cancer treatment, cytostaticity and cytotoxicity, will be discussed.
Collapse
Affiliation(s)
- Jan de Weille
- Institut des Neurosciences de Montpellier, U1051 INSERM, 80 rue Augustin Fliche, 34295 Montpellier Cedex 05, France.
| | | | | |
Collapse
|
30
|
Rao S, Ge S, Shelly M. Centrosome positioning and primary cilia assembly orchestrate neuronal development. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/s11515-012-1231-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
31
|
Mumert M, Dubuc A, Wu X, Northcott PA, Chin SS, Pedone CA, Taylor MD, Fults DW. Functional genomics identifies drivers of medulloblastoma dissemination. Cancer Res 2012; 72:4944-53. [PMID: 22875024 DOI: 10.1158/0008-5472.can-12-1629] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Medulloblastomas are malignant brain tumors that arise in the cerebellum in children and disseminate via the cerebrospinal fluid to the leptomeningeal spaces of the brain and spinal cord. Challenged by the poor prognosis for patients with metastatic dissemination, pediatric oncologists have developed aggressive treatment protocols, combining surgery, craniospinal radiation, and high-dose chemotherapy, that often cause disabling neurotoxic effects in long-term survivors. Insights into the genetic control of medulloblastoma dissemination have come from transposon insertion mutagenesis studies. Mobilizing the Sleeping Beauty transposon in cerebellar neural progenitor cells caused widespread dissemination of typically nonmetastatic medulloblastomas in Patched(+/-) mice, in which Shh signaling is hyperactive. Candidate metastasis genes were identified by sequencing the insertion sites and then mapping these sequences back to the mouse genome. To determine whether genes located at transposon insertion sites directly caused medulloblastomas to disseminate, we overexpressed candidate genes in Nestin(+) neural progenitors in the cerebella of mice by retroviral transfer in combination with Shh. We show here that ectopic expression of Eras, Lhx1, Ccrk, and Akt shifted the in vivo growth characteristics of Shh-induced medulloblastomas from a localized pattern to a disseminated pattern in which tumor cells seeded the leptomeningeal spaces of the brain and spinal cord.
Collapse
Affiliation(s)
- Michael Mumert
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The original hedgehog (hh) gene was found in Drosophila and named for the appearance of a mutant phenotype which causes an embryo to be covered with pointy denticles, thus resembling a hedgehog. The hedgehog family consists of sonic hedgehog (Shh), desert hedgehog (Dhh), and Indian hedgehog (Ihh). Shh is found in vertebrates and the best studied ligand of the hedgehog signaling pathway (Gilbert, 2000). It plays an important role in regulating vertebrate organogenesis, such as in the growth of digits on limbs and organization of the brain, and earlier studies also show that it is important during retinal development (for a review, see Amato et al., 2004). Hedgehog expression drives waves of neurogenesis in animal retina, although genetic programs that control its expression are poorly elucidated. Recently, a novel transcriptional cascade which involves the atonal and Iroquois gene family was proposed in the regulation of hedgehog waves during vertebrate retinal development (Choy et al., 2010). This chapter will focus on Shh by addressing its signaling mechanisms and roles in vertebrate eye development, as well as a novel finding in retinogenesis.
Collapse
|
33
|
McNeill B, Perez-Iratxeta C, Mazerolle C, Furimsky M, Mishina Y, Andrade-Navarro MA, Wallace VA. Comparative genomics identification of a novel set of temporally regulated hedgehog target genes in the retina. Mol Cell Neurosci 2012; 49:333-40. [PMID: 22281533 DOI: 10.1016/j.mcn.2011.12.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 11/07/2011] [Accepted: 12/15/2011] [Indexed: 11/26/2022] Open
Abstract
The hedgehog (Hh) signaling pathway is involved in numerous developmental and adult processes with many links to cancer. In vertebrates, the activity of the Hh pathway is mediated primarily through three Gli transcription factors (Gli1, 2 and 3) that can serve as transcriptional activators or repressors. The identification of Gli target genes is essential for the understanding of the Hh-mediated processes. We used a comparative genomics approach using the mouse and human genomes to identify 390 genes that contained conserved Gli binding sites. RT-qPCR validation of 46 target genes in E14.5 and P0.5 retinal explants revealed that Hh pathway activation resulted in the modulation of 30 of these targets, 25 of which demonstrated a temporal regulation. Further validation revealed that the expression of Bok, FoxA1, Sox8 and Wnt7a was dependent upon Sonic Hh (Shh) signaling in the retina and their regulation is under positive and negative controls by Gli2 and Gli3, respectively. We also show using chromatin immunoprecipitation that Gli2 binds to the Sox8 promoter, suggesting that Sox8 is an Hh-dependent direct target of Gli2. Finally, we demonstrate that the Hh pathway also modulates the expression of Sox9 and Sox10, which together with Sox8 make up the SoxE group. Previously, it has been shown that Hh and SoxE group genes promote Müller glial cell development in the retina. Our data are consistent with the possibility for a role of SoxE group genes downstream of Hh signaling on Müller cell development.
Collapse
Affiliation(s)
- Brian McNeill
- Vision Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | | | | | | | | | | | | |
Collapse
|
34
|
Ahmed AI, Shtaya AB, Zaben MJ, Owens EV, Kiecker C, Gray WP. Endogenous GFAP-positive neural stem/progenitor cells in the postnatal mouse cortex are activated following traumatic brain injury. J Neurotrauma 2011; 29:828-42. [PMID: 21895532 DOI: 10.1089/neu.2011.1923] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interest in promoting regeneration of the injured nervous system has recently turned toward the use of endogenous stem cells. Elucidating cues involved in driving these precursor cells out of quiescence following injury, and the signals that drive them toward neuronal and glial lineages, will help to harness these cells for repair. Using a biomechanically validated in vitro organotypic stretch injury model, cortico-hippocampal slices from postnatal mice were cultured and a stretch injury equivalent to a severe traumatic brain injury (TBI) applied. In uninjured cortex, proliferative potential under in vitro conditions is virtually absent in older slices (equivalent postnatal day 15 compared to 8). However, following a severe stretch injury, this potential is restored in injured outer cortex. Using slices from mice expressing a fluorescent reporter on the human glial fibrillary acidic protein (GFAP) promoter, we show that GFAP+ cells account for the majority of proliferating neurospheres formed, and that these cells are likely to arise from the cortical parenchyma and not from the subventricular zone. Moreover, we provide evidence for a correlation between upregulation of sonic hedgehog signaling, a pathway known to regulate stem cell proliferation, and this restoration of regenerative potential following TBI. Our results indicate that a source of quiescent endogenous stem cells residing in the cortex and subcortical tissue proliferate in vitro following TBI. Moreover, these proliferating cells are multipotent and are derived mostly from GFAP-expressing cells. This raises the possibility of using this endogenous source of stem cells for repair following TBI.
Collapse
Affiliation(s)
- Aminul I Ahmed
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | | | | | | | | |
Collapse
|
35
|
Trisomy 12 and elevated GLI1 and PTCH1 transcript levels are biomarkers for Hedgehog-inhibitor responsiveness in CLL. Blood 2011; 119:997-1007. [PMID: 22130798 DOI: 10.1182/blood-2011-06-359075] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hedgehog (HH) signaling is activated in various lymphoid malignancies, but conflicting results exist about its role in chronic lymphocytic leukemia (CLL). Here, we demonstrate that the expression of essential HH pathway components like GLI1, PTCH1, and the HH ligands is highly diverse in CLL. A subset of 36.7% of 60 tested CLL samples responded to all 3 SMOOTHENED (SMO) inhibitors, whereas 40% were completely resistant. Responsiveness correlated with elevated GLI1 and PTCH1 transcript levels and the presence of trisomy 12, whereas no other karyotype correlated with responsiveness. All trisomy 12 CLLs displayed constitutive HH pathway activation driven by autocrine DESERT HH (DHH) ligand secretion, which could be blocked by the HH-blocking Ab 5E1. Cocultures with DHH-expressing BM stromal cells reduced sensitivity of CLLs to SMO-inhibitor treatment by activation of noncanonical ERK phosphorylation directly downstream of the PTCH1 receptor without involvement of SMO and could be overcome by the HH-blocking Ab 5E1 or a combination of SMO and ERK inhibitors. Our results demonstrate that the HH-signaling pathway is an interesting therapeutic target for a subset of patients with CLL, characterized by high GLI1 and PTCH1 transcript levels, and all patients with trisomy 12 and indicate HH-blocking Abs to be favorable over SMO inhibitors in overcoming stroma-mediated protective effects.
Collapse
|
36
|
Abstract
The Hedgehog (Hh) pathway is a major regulator of many fundamental processes in vertebrate embryonic development including stem cell maintenance, cell differentiation, tissue polarity and cell proliferation. Constitutive activation of the Hh pathway leading to tumorigenesis is seen in basal cell carcinomas and medulloblastoma. A variety of other human cancers, including brain, gastrointestinal, lung, breast and prostate cancers, also demonstrate inappropriate activation of this pathway. Paracrine Hh signaling from the tumor to the surrounding stroma was recently shown to promote tumorigenesis. This pathway has also been shown to regulate proliferation of cancer stem cells and to increase tumor invasiveness. Targeted inhibition of Hh signaling may be effective in the treatment and prevention of many types of human cancers. The discovery and synthesis of specific Hh pathway inhibitors have significant clinical implications in novel cancer therapeutics. Several synthetic Hh antagonists are now available, several of which are undergoing clinical evaluation. The orally available compound, GDC-0449, is the farthest along in clinical development. Initial clinical trials in basal cell carcinoma and treatment of select patients with medulloblastoma have shown good efficacy and safety. We review the molecular basis of Hh signaling, the current understanding of pathway activation in different types of human cancers and we discuss the clinical development of Hh pathway inhibitors in human cancer therapy.
Collapse
Affiliation(s)
- Sachin Gupta
- Wayne State University, Karmanos Cancer Institute, Detroit, MI, USA
| | | | | |
Collapse
|
37
|
Zhang Y, Hao H, Zhao S, Liu Q, Yuan Q, Ni S, Wang F, Liu S, Wang L, Hao A. Downregulation of GRIM-19 promotes growth and migration of human glioma cells. Cancer Sci 2011; 102:1991-9. [PMID: 21827581 DOI: 10.1111/j.1349-7006.2011.02059.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
It has become increasingly clear that there are notable parallels between normal development and tumorigenesis. Glioma is a classic model that links between tumorigenesis and development. We evaluated the expression of GRIM-19, a novel gene essential for normal development, in various grades of gliomas and several human glioma cell lines. We showed that GRIM-19 mRNA and protein expression were markedly lower in gliomas than in control brain tissues and negatively correlated with the malignancy of gliomas. Downregulation of GRIM-19 in glioma cells significantly enhanced cell proliferation and migration, whereas overexpression of GRIM-19 showed the opposite effects. We also showed that the activation of signal transducer and activator of transcription 3 (STAT3) and the expression of many STAT3-dependent genes were regulated by the expression of GRIM-19. In addition, GRIM-19 exerted its role probably through the non-STAT3 signaling pathway. Collectively, our data suggest that most gliomas expressed GRIM-19 at low levels, which may play a major role in tumorigenesis in the brain.
Collapse
Affiliation(s)
- Yanmin Zhang
- Key Laboratory of Ministry of Education for Experimental Teratology, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Balbuena J, Pachon G, Lopez-Torrents G, Aran JM, Castresana JS, Petriz J. ABCG2 is required to control the sonic hedgehog pathway in side population cells with stem-like properties. Cytometry A 2011; 79:672-83. [PMID: 21774076 DOI: 10.1002/cyto.a.21103] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 05/23/2011] [Accepted: 06/15/2011] [Indexed: 11/06/2022]
Abstract
The Sonic Hedgehog (Hh) pathway has been implicated in the maintenance of stem or progenitor cells in many adult tissues. Importantly, abnormal Hh pathway activation is also associated with initiation of neoplasia, but its role in tumor growth is still unclear. Here, we demonstrate that cyclopamine, a plant-derived alkaloid product used to inhibit the Hh signaling pathway, reduces the Side Population (SP) obtained by Hoechst 33342 (Ho342) dye measurements. In addition, cyclopamine is able to modulate, along with oxysterols and other products, the ABCG2 transporter by increasing Ho342 and mitoxantrone uptake. Therefore, if the SP is solely measured as a Ho342 dye extruding fraction, this may be significantly modulated by the inhibition of ABCG2 transport fraction, independently from the action of cyclopamine on the Hh pathway. Our results indicate that ABCG2 may act in the upstream regulation of the Hh signaling pathway to protect the stemness of the SP compartment, giving support to the cancer stem cell hypothesis and suggesting that ABCG2 is not only critical for increased resistance to anticancer agents.
Collapse
Affiliation(s)
- Jana Balbuena
- Brain Tumor Biology Unit, CIFA, University of Navarra School of Sciences, Pamplona, Spain
| | | | | | | | | | | |
Collapse
|
39
|
Marcucio RS, Young NM, Hu D, Hallgrimsson B. Mechanisms that underlie co-variation of the brain and face. Genesis 2011; 49:177-89. [PMID: 21381182 PMCID: PMC3086711 DOI: 10.1002/dvg.20710] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 12/15/2010] [Accepted: 12/23/2010] [Indexed: 12/11/2022]
Abstract
The effect of the brain on the morphology of the face has long been recognized in both evolutionary biology and clinical medicine. In this work, we describe factors that are active between the development of the brain and face and how these might impact craniofacial variation. First, there is the physical influence of the brain, which contributes to overall growth and morphology of the face through direct structural interactions. Second, there is the molecular influence of the brain, which signals to facial tissues to establish signaling centers that regulate patterned growth. Importantly, subtle alterations to these physical or molecular interactions may contribute to both normal and abnormal variation. These interactions are therefore critical to our understanding of how a diversity of facial morphologies can be generated both within species and across evolutionary time.
Collapse
Affiliation(s)
- Ralph S Marcucio
- University of California, San Francisco, Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, UCSF, San Francisco General Hospital, San Francisco, California 94110, USA.
| | | | | | | |
Collapse
|
40
|
Logan CV, Abdel-Hamed Z, Johnson CA. Molecular genetics and pathogenic mechanisms for the severe ciliopathies: insights into neurodevelopment and pathogenesis of neural tube defects. Mol Neurobiol 2010; 43:12-26. [PMID: 21110233 DOI: 10.1007/s12035-010-8154-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 11/16/2010] [Indexed: 01/05/2023]
Abstract
Meckel-Gruber syndrome (MKS) is a severe autosomal recessively inherited disorder characterized by developmental defects of the central nervous system that comprise neural tube defects that most commonly present as occipital encephalocele. MKS is considered to be the most common syndromic form of neural tube defect. MKS is genetically heterogeneous with six known disease genes: MKS1, MKS2/TMEM216, MKS3/TMEM67, RPGRIP1L, CEP290, and CC2D2A with the encoded proteins all implicated in the correct function of primary cilia. Primary cilia are microtubule-based organelles that project from the apical surface of most epithelial cell types. Recent progress has implicated the involvement of cilia in the Wnt and Shh signaling pathways and has led to an understanding of their role in normal mammalian neurodevelopment. The aim of this review is to provide an overview of the molecular genetics of the human disorder, and to assess recent insights into the etiology and molecular cell biology of severe ciliopathies from mammalian animal models of MKS.
Collapse
Affiliation(s)
- Clare V Logan
- Section of Ophthalmology and Neurosciences, Wellcome Trust Brenner Building, Leeds Institute of Molecular Medicine, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | | | | |
Collapse
|
41
|
Gianakopoulos PJ, Mehta V, Voronova A, Cao Y, Yao Z, Coutu J, Wang X, Waddington MS, Tapscott SJ, Skerjanc IS. MyoD directly up-regulates premyogenic mesoderm factors during induction of skeletal myogenesis in stem cells. J Biol Chem 2010; 286:2517-25. [PMID: 21078671 DOI: 10.1074/jbc.m110.163709] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gain- and loss-of-function experiments have illustrated that the family of myogenic regulatory factors is necessary and sufficient for the formation of skeletal muscle. Furthermore, MyoD required cellular aggregation to induce myogenesis in P19 embryonal carcinoma stem cells. To determine the mechanism by which stem cells can be directed into skeletal muscle, a time course of P19 cell differentiation was examined in the presence and absence of exogenous MyoD. By quantitative PCR, the first MyoD up-regulated transcripts were the premyogenic mesoderm factors Meox1, Pax7, Six1, and Eya2 on day 4 of differentiation. Subsequently, the myoblast markers myogenin, MEF2C, and Myf5 were up-regulated, leading to skeletal myogenesis. These results were corroborated by Western blot analysis, showing up-regulation of Pax3, Six1, and MEF2C proteins, prior to myogenin protein expression. To determine at what stage a dominant-negative MyoD/EnR mutant could inhibit myogenesis, stable cell lines were created and examined. Interestingly, the premyogenic mesoderm factors, Meox1, Pax3/7, Six1, Eya2, and Foxc1, were down-regulated, and as expected, skeletal myogenesis was abolished. Finally, to identify direct targets of MyoD in this system, chromatin immunoprecipitation experiments were performed. MyoD was observed associated with regulatory regions of Meox1, Pax3/7, Six1, Eya2, and myogenin genes. Taken together, MyoD directs stem cells into the skeletal muscle lineage by binding and activating the expression of premyogenic mesoderm genes, prior to activating myoblast genes.
Collapse
Affiliation(s)
- Peter J Gianakopoulos
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Garcia ADR, Petrova R, Eng L, Joyner AL. Sonic hedgehog regulates discrete populations of astrocytes in the adult mouse forebrain. J Neurosci 2010; 30:13597-608. [PMID: 20943901 PMCID: PMC2966838 DOI: 10.1523/jneurosci.0830-10.2010] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Revised: 06/29/2010] [Accepted: 07/29/2010] [Indexed: 01/02/2023] Open
Abstract
Astrocytes are an essential component of the CNS, and recent evidence points to an increasing diversity of their functions. Identifying molecular pathways that mediate distinct astrocyte functions, is key to understanding how the nervous system operates in the intact and pathological states. In this study, we demonstrate that the Hedgehog (Hh) pathway, well known for its roles in the developing CNS, is active in astrocytes of the mature mouse forebrain in vivo. Using multiple genetic approaches, we show that regionally distinct subsets of astrocytes receive Hh signaling, indicating a molecular diversity between specific astrocyte populations. Furthermore, we identified neurons as a source of Sonic hedgehog (Shh) in the adult forebrain, suggesting that Shh signaling is involved in neuron-astrocyte communication. Attenuation of Shh signaling in postnatal astrocytes by targeted removal of Smoothened, an obligate Shh coreceptor, resulted in upregulation of GFAP and cellular hypertrophy specifically in astrocyte populations regulated by Shh signaling. Collectively, our findings demonstrate a role for neuron-derived Shh in regulating specific populations of differentiated astrocytes.
Collapse
Affiliation(s)
- A. Denise R. Garcia
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, Sloan-Kettering Institute, New York, New York 10065
| | - Ralitsa Petrova
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, Sloan-Kettering Institute, New York, New York 10065
- Biochemistry, Cell, and Molecular Biology Program, Weill Graduate School of Medical Sciences, Cornell University, New York, New York 10065
| | - Liane Eng
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, Sloan-Kettering Institute, New York, New York 10065
| | - Alexandra L. Joyner
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, Sloan-Kettering Institute, New York, New York 10065
| |
Collapse
|
43
|
Chen Y, Peng C, Sullivan C, Li D, Li S. Critical molecular pathways in cancer stem cells of chronic myeloid leukemia. Leukemia 2010; 24:1545-54. [PMID: 20574455 PMCID: PMC3130198 DOI: 10.1038/leu.2010.143] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 04/27/2010] [Accepted: 05/26/2010] [Indexed: 01/17/2023]
Abstract
Inhibition of BCR-ABL with kinase inhibitors in the treatment of Philadelphia-positive (Ph(+)) chronic myeloid leukemia (CML) is highly effective in controlling but not curing the disease. This is largely due to the inability of these kinase inhibitors to kill leukemia stem cells (LSCs) responsible for disease relapse. This stem cell resistance is not associated with the BCR-ABL kinase domain mutations resistant to kinase inhibitors. Development of curative therapies for CML requires the identification of crucial molecular pathways responsible for the survival and self-renewal of LSCs. In this review, we will discuss our current understanding of these crucial molecular pathways in LSCs and the available therapeutic strategies for targeting these stem cells in CML.
Collapse
Affiliation(s)
- Yaoyu Chen
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Cong Peng
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Con Sullivan
- Department of Cancer Research, Maine Institute for Human Genetics and Health, 246 Sylvan Road, Bangor, ME 04401, USA
| | - Dongguang Li
- School of Computer and Security Science, Edith Cowan University, 2 Bradford Street, Mount Lawley, WA 6050, Australia
| | - Shaoguang Li
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| |
Collapse
|
44
|
Coon V, Laukert T, Pedone CA, Laterra J, Kim KJ, Fults DW. Molecular therapy targeting Sonic hedgehog and hepatocyte growth factor signaling in a mouse model of medulloblastoma. Mol Cancer Ther 2010; 9:2627-36. [PMID: 20807782 PMCID: PMC2937075 DOI: 10.1158/1535-7163.mct-10-0486] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The use of genetically engineered mice has provided insights into the molecular pathogenesis of the pediatric brain tumor medulloblastoma and revealed promising therapeutic targets. Ectopic expression of Sonic hedgehog (Shh) in cerebellar neural progenitor cells induces medulloblastomas in mice, and coexpression of hepatocyte growth factor (HGF) enhances Shh-induced tumor formation. To determine whether Shh + HGF-driven medulloblastomas were responsive to Shh signaling blockade and whether treatment response could be enhanced by combination therapy targeting both HGF and Shh signaling pathways, we carried out a survival study in mice. We induced medulloblastomas by retrovirus-mediated expression of Shh and HGF, after which we treated the mice systemically with (a) HGF-neutralizing monoclonal antibody L2G7, (b) Shh signaling inhibitor cyclopamine, (c) Shh-neutralizing monoclonal antibody 5E1, (d) L2G7 + cyclopamine, or (e) L2G7 + 5E1. We report that monotherapy targeting either HGF signaling or Shh signaling prolonged survival and that anti-HGF therapy had a more durable response than Shh-targeted therapy. The effect of L2G7 + 5E1 combination therapy on cumulative survival was equivalent to that of L2G7 monotherapy and that of L2G7 + cyclopamine therapy was worse. The principal mechanism by which Shh- and HGF-targeted therapies inhibited tumor growth was a potent apoptotic death response in tumor cells, supplemented by a weaker suppressive effect on proliferation. Our observation that combination therapy either failed to improve or even reduced survival in mice bearing Shh + HGF-induced medulloblastomas compared with monotherapy underscores the importance of preclinical testing of molecular-targeted therapies in animal models of tumors in which the targeted pathways are known to be active.
Collapse
Affiliation(s)
- Valerie Coon
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Tamara Laukert
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Carolyn A. Pedone
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - John Laterra
- Department of Neurology, The Kennedy Krieger Institute, and Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - K. Jin Kim
- Galaxy Biotech, LLC; Mountain View, CA, USA
| | - Daniel W. Fults
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
45
|
Bragina O, Sergejeva S, Serg M, Zarkovsky T, Maloverjan A, Kogerman P, Zarkovsky A. Smoothened agonist augments proliferation and survival of neural cells. Neurosci Lett 2010; 482:81-5. [PMID: 20600593 DOI: 10.1016/j.neulet.2010.06.068] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 06/22/2010] [Accepted: 06/23/2010] [Indexed: 11/26/2022]
Abstract
Sonic hedgehog signaling pathway is important in developmental processes like dorsoventral neural tube patterning, neural stem cell proliferation and neuronal and glial cell survival. Shh is also implicated in the regulation of the adult hippocampal neurogenesis. Recently, nonpeptidyl Smoothened activators of the Shh pathway have been identified. The aim of this study was to investigate the effects of chlorobenzothiophene-containing molecule, Smo agonist (SAG), which has been shown to activate Shh signaling pathway, in neurogenesis and neuronal survival in in vitro and in vivo models. Our in vitro experiments showed that SAG induces increased expression of Gli1 mRNA, transcriptional target and mediator of Shh signal. In vitro experiments also demonstrated that SAG in low-nanomolar concentrations induces proliferation of neuronal and glial precursors without affecting the differentiation pattern of newly produced cells. In contrast to Shh, SAG did not induce neurotoxicity in neuronal cultures. The SAG and Shh treatment also promoted the survival of newly generated neural cells in the dentate gyrus after their intracerebroventricular administration to adult rats. We propose that SAG and similar compounds represent attractive molecules to be developed for treatment of disorders where stimulation of the generation and survival of new neural cells would be beneficial.
Collapse
Affiliation(s)
- Olga Bragina
- Institute of Clinical Medicine, Tallinn University of Technology, Estonia
| | | | | | | | | | | | | |
Collapse
|
46
|
New developments in the discovery of small molecule Hedgehog pathway antagonists. Curr Opin Chem Biol 2010; 14:428-35. [DOI: 10.1016/j.cbpa.2010.03.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 03/19/2010] [Accepted: 03/21/2010] [Indexed: 01/14/2023]
|
47
|
Sullivan JP, Minna JD, Shay JW. Evidence for self-renewing lung cancer stem cells and their implications in tumor initiation, progression, and targeted therapy. Cancer Metastasis Rev 2010; 29:61-72. [PMID: 20094757 DOI: 10.1007/s10555-010-9216-5] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The discovery of rare tumor cells with stem cell features first in leukemia and later in solid tumors has emerged as an important area in cancer research. It has been determined that these stem-like tumor cells, termed cancer stem cells, are the primary cellular component within a tumor that drives disease progression and metastasis. In addition to their stem-like ability to self-renew and differentiate, cancer stem cells are also enriched in cells resistant to conventional radiation therapy and to chemotherapy. The immediate implications of this new tumor growth paradigm not only require a re-evaluation of how tumors are initiated, but also on how tumors should be monitored and treated. However, despite the relatively rapid pace of cancer stem cell research in solid tumors such as breast, brain, and colon cancers, similar progress in lung cancer remains hampered in part due to an incomplete understanding of lung epithelial stem cell hierarchy and the complex heterogeneity of the disease. In this review, we provide a critical summary of what is known about the role of normal and malignant lung stem cells in tumor development, the progress in characterizing lung cancer stem cells and the potential for therapeutically targeting pathways of lung cancer stem cell self-renewal.
Collapse
Affiliation(s)
- James P Sullivan
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| | | | | |
Collapse
|
48
|
Abstract
Hedgehog signaling is activated in a variety of solid tumors and hematologic malignancies by point mutations in hedgehog pathway members or autocrine or paracrine ligand secretion. Several hedgehog inhibitors were developed to block hedgehog pathway activity on the level of the activating receptor, Smoothened (Smo). GDC-0449 is the first systemic Smo-inhibitor entering clinical trials. It was successfully tested in a phase-I clinical trial demonstrating good pharmacodynamic (PD) and pharmacokinetic (PK) properties and showing objective response and clinical benefit in several patients with basal cell carcinoma.
Collapse
Affiliation(s)
- Christine Dierks
- Department of Hematology and Oncology, Freiburg University Medical Center, Hugstetterstrasse. 55, 79106, Freiburg, Germany.
| |
Collapse
|
49
|
Planarian Hedgehog/Patched establishes anterior-posterior polarity by regulating Wnt signaling. Proc Natl Acad Sci U S A 2009; 106:22329-34. [PMID: 20018728 DOI: 10.1073/pnas.0907464106] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite long-standing interest, the molecular mechanisms underlying the establishment of anterior-posterior (AP) polarity remain among the unsolved mysteries in metazoans. In the planarians (a family of flatworms), canonical Wnt/beta-catenin signaling is required for posterior specification, as it is in many animals. However, the molecular mechanisms regulating the posterior-specific induction of Wnt genes according to the AP polarity have remained unclear. Here, we demonstrate that Hedgehog (Hh) signaling is responsible for the establishment of AP polarity via its regulation of the transcription of Wnt family genes during planarian regeneration. We found that RNAi gene knockdown of Dugesia japonica patched (Djptc) caused ectopic tail formation in the anterior blastema of body fragments, resulting in bipolar-tails regeneration. In contrast, RNAi of hedgehog (Djhh) and gli (Djgli) caused bipolar-heads regeneration. We show that Patched-mediated Hh signaling was crucial for posterior specification, which is established by regulating the transcription of Wnt genes via downstream Gli activity. Moreover, differentiated cells were responsible for the posterior specification of undifferentiated stem cells through Wnt/beta-catenin signaling. Surprisingly, Djhh was expressed in neural cells all along the ventral nerve cords (along the AP axis), but not in the posterior blastema of body fragments, where the expression of Wnt genes was induced for posteriorization. We therefore propose that Hh signals direct head or tail regeneration according to the AP polarity, which is established by Hh signaling activity along the body's preexisting nervous system.
Collapse
|
50
|
Tremblay MR, Nesler M, Weatherhead R, Castro AC. Recent patents for Hedgehog pathway inhibitors for the treatment of malignancy. Expert Opin Ther Pat 2009; 19:1039-56. [PMID: 19505195 DOI: 10.1517/13543770903008551] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND There is increasing evidence suggesting that blocking aberrant Hedgehog (Hh) signaling can be a novel therapeutic avenue for the treatment of cancer. During the past decade, efforts from academic and industrial groups have led to the discovery of a variety of Hh pathway inhibitors. OBJECTIVE This review covers the patent literature related to Hh pathway inhibitors for the treatment of proliferative diseases, regardless of their modes of action. METHODS A comprehensive survey of the patent literature since 1999 is presented. RESULTS/CONCLUSION Most reported Hh pathway inhibitors act on the key signaling transducer Smoothened (SMO). Screening of compound libraries using reporter and binding assays have identified a broad diversity of chemical structures that interact with SMO. These screening approaches, followed by conventional medicinal chemistry, have delivered important clinical drug candidates, such as GDC-0449 and XL-139. In addition, modification of the naturally occurring Veratrum alkaloid cyclopamine has resulted in various active analogues, including clinical drug candidate IPI-926. Although there are recent scientific literature reports of small molecules acting downstream of SMO, there is limited patent literature on this mode of Hh pathway inhibition.
Collapse
|