1
|
Ascanelli C, Dahir R, Wilson CH. Manipulating Myc for reparative regeneration. Front Cell Dev Biol 2024; 12:1357589. [PMID: 38577503 PMCID: PMC10991803 DOI: 10.3389/fcell.2024.1357589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/15/2024] [Indexed: 04/06/2024] Open
Abstract
The Myc family of proto-oncogenes is a key node for the signal transduction of external pro-proliferative signals to the cellular processes required for development, tissue homoeostasis maintenance, and regeneration across evolution. The tight regulation of Myc synthesis and activity is essential for restricting its oncogenic potential. In this review, we highlight the central role that Myc plays in regeneration across the animal kingdom (from Cnidaria to echinoderms to Chordata) and how Myc could be employed to unlock the regenerative potential of non-regenerative tissues in humans for therapeutic purposes. Mastering the fine balance of harnessing the ability of Myc to promote transcription without triggering oncogenesis may open the door to many exciting opportunities for therapeutic development across a wide array of diseases.
Collapse
Affiliation(s)
| | | | - Catherine H. Wilson
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
2
|
Purhonen J, Klefström J, Kallijärvi J. MYC-an emerging player in mitochondrial diseases. Front Cell Dev Biol 2023; 11:1257651. [PMID: 37731815 PMCID: PMC10507175 DOI: 10.3389/fcell.2023.1257651] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
The mitochondrion is a major hub of cellular metabolism and involved directly or indirectly in almost all biological processes of the cell. In mitochondrial diseases, compromised respiratory electron transfer and oxidative phosphorylation (OXPHOS) lead to compensatory rewiring of metabolism with resemblance to the Warburg-like metabolic state of cancer cells. The transcription factor MYC (or c-MYC) is a major regulator of metabolic rewiring in cancer, stimulating glycolysis, nucleotide biosynthesis, and glutamine utilization, which are known or predicted to be affected also in mitochondrial diseases. Albeit not widely acknowledged thus far, several cell and mouse models of mitochondrial disease show upregulation of MYC and/or its typical transcriptional signatures. Moreover, gene expression and metabolite-level changes associated with mitochondrial integrated stress response (mt-ISR) show remarkable overlap with those of MYC overexpression. In addition to being a metabolic regulator, MYC promotes cellular proliferation and modifies the cell cycle kinetics and, especially at high expression levels, promotes replication stress and genomic instability, and sensitizes cells to apoptosis. Because cell proliferation requires energy and doubling of the cellular biomass, replicating cells should be particularly sensitive to defective OXPHOS. On the other hand, OXPHOS-defective replicating cells are predicted to be especially vulnerable to high levels of MYC as it facilitates evasion of metabolic checkpoints and accelerates cell cycle progression. Indeed, a few recent studies demonstrate cell cycle defects and nuclear DNA damage in OXPHOS deficiency. Here, we give an overview of key mitochondria-dependent metabolic pathways known to be regulated by MYC, review the current literature on MYC expression in mitochondrial diseases, and speculate how its upregulation may be triggered by OXPHOS deficiency and what implications this has for the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Janne Purhonen
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juha Klefström
- Finnish Cancer Institute, FICAN South Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine, Medical Faculty, University of Helsinki, Helsinki, Finland
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Jukka Kallijärvi
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
Dash SN, Patnaik L. Flight for fish in drug discovery: a review of zebrafish-based screening of molecules. Biol Lett 2023; 19:20220541. [PMID: 37528729 PMCID: PMC10394424 DOI: 10.1098/rsbl.2022.0541] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 07/13/2023] [Indexed: 08/03/2023] Open
Abstract
Human disease and biological practices are modelled in zebrafish (Danio rerio) at various phases of drug development as well as toxicity evaluation. The zebrafish is ideal for in vivo pathological research and high-resolution investigation of disease progress. Zebrafish has an advantage over other mammalian models, it is cost-effective, it has external development and embryo transparency, easy to apply genetic manipulations, and open to both forward and reverse genetic techniques. Drug screening in zebrafish is suitable for target identification, illness modelling, high-throughput screening of compounds for inhibition or prevention of disease phenotypes and developing new drugs. Several drugs that have recently entered the clinic or clinical trials have their origins in zebrafish. The sophisticated screening methods used in zebrafish models are expected to play a significant role in advancing drug development programmes. This review highlights the current developments in drug discovery processes, including understanding the action of drugs in the context of disease and screening novel candidates in neurological diseases, cardiovascular diseases, glomerulopathies and cancer. Additionally, it summarizes the current techniques and approaches for the selection of small molecules and current technical limitations on the execution of zebrafish drug screening tests.
Collapse
Affiliation(s)
- Surjya Narayan Dash
- Institute of Biotechnology, Biocenter 2. Viikinkaari, University of Helsinki, Viikinkaari 5D, 00790 Helsinki, Finland
| | - Lipika Patnaik
- Environmental Science Laboratory, Department of Zoology, COE in Environment and Public Health, Ravenshaw University, Cuttack 751003, Odisha, India
| |
Collapse
|
4
|
Mitra S, Sharma P, Kaur S, Khursheed MA, Gupta S, Chaudhary M, Kurup AJ, Ramachandran R. Dual regulation of lin28a by Myc is necessary during zebrafish retina regeneration. J Cell Biol 2019; 218:489-507. [PMID: 30606747 PMCID: PMC6363449 DOI: 10.1083/jcb.201802113] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 08/31/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023] Open
Abstract
Cellular reprogramming leading to induction of Muller glia-derived progenitor cells (MGPCs) with stem cell characteristics is essential for zebrafish retina regeneration. Although several regeneration-specific genes are characterized, the significance of MGPC-associated Mycb induction remains unknown. Here, we show that early expression of Mycb induces expression of genes like ascl1a, a known activator of lin28a in MGPCs. Notably, mycb is simultaneously activated by Ascl1a and repressed by Insm1a in regenerating retina. Here, we unravel a dual role of Mycb in lin28a expression, both as an activator through Ascl1a in MGPCs and a repressor in combination with Hdac1 in neighboring cells. Myc inhibition reduces the number of MGPCs and abolishes normal regeneration. Myc in collaboration with Hdac1 inhibits her4.1, an effector of Delta-Notch signaling. Further, we also show the repressive role of Delta-Notch signaling on lin28a expression in post-injured retina. Our studies reveal mechanistic understanding of Myc pathway during zebrafish retina regeneration, which could pave way for therapeutic intervention during mammalian retina regeneration.
Collapse
Affiliation(s)
- Soumitra Mitra
- Indian Institute of Science Education and Research, Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, India
| | - Poonam Sharma
- Indian Institute of Science Education and Research, Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, India
| | - Simran Kaur
- Indian Institute of Science Education and Research, Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, India
| | - Mohammad Anwar Khursheed
- Indian Institute of Science Education and Research, Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, India
| | - Shivangi Gupta
- Indian Institute of Science Education and Research, Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, India
| | - Mansi Chaudhary
- Indian Institute of Science Education and Research, Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, India
| | - Akshai J Kurup
- Indian Institute of Science Education and Research, Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, India
| | - Rajesh Ramachandran
- Indian Institute of Science Education and Research, Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, India
| |
Collapse
|
5
|
Jiang Y, Han K, Chen S, Wang Y, Zhang Z. Isolation, characterization, and expression of proto-oncogene cMyc in large yellow croaker Larimichthys crocea. FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:1443-1461. [PMID: 28550411 DOI: 10.1007/s10695-017-0384-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 05/08/2017] [Indexed: 06/07/2023]
Abstract
cMyc is a vital transcription factor that involves in the regulation of cell proliferation, growth, differentiation, and apoptosis. In the present study, cMyc in Larimichthys crocea (Lc-cMyc) was cloned and analyzed for investigating its function. The full-length cDNA of Lc-cMyc was 2089 bp encoding a 440-amino-acid protein (Lc-cMyc). Lc-cMyc had the characteristic helix-loop-helix-leucine-zipper (HLH-LZ) DNA-binding domain and highly conservative in evolution. The expression of Lc-cMyc was detected by quantitative real-time PCR (qRT-PCR) and in situ hybridization, respectively. In tissues, the gender differences of Lc-cMyc expression existed only in gonad and Lc-cMyc was extremely significantly expressed in ovary with the highest level in 635-dph ovary, especially in stages II (late) and III (early) oocytes. A certain degree of expression was examined in head kidney of both sexes and testis with high expression in spermatocyte. In embryos, Lc-cMyc was expressed at all embryonic stages. In early embryogenesis (from two-cell stage to mutiple-cell stage), Lc-cMyc was expressed very highly with a peak at two-cell stage. In late embryogenesis (from blastula stage to 1-day-post-hatching stage), the high expression of Lc-cMyc was detected as the following order: 1-day-post-hatching stage > pre-hatching stage > the appearance-of-optic-vesicles stage = mutiple-cell stage > beginning-of-heart-pulsation stage. The distribution of Lc-cMyc concentrated gradually in the back of embryos until in the head. In conclusion, the spatio-temporal expression patterns of Lc-cMyc indicated an essential role in oogenesis and embryogenesis and contributed to insight into the molecular mechanisms of regulating pluripotency in large yellow croaker.
Collapse
Affiliation(s)
- Yonghua Jiang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen, 361021, China
- State Key Laboratory of Large Yellow Croaker Breeding, Ningde Fufa Fisheries Company Limited, Ningde, 352103, China
| | - Kunhuang Han
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen, 361021, China
- State Key Laboratory of Large Yellow Croaker Breeding, Ningde Fufa Fisheries Company Limited, Ningde, 352103, China
| | - Shihai Chen
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Yilei Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen, 361021, China.
- State Key Laboratory of Large Yellow Croaker Breeding, Ningde Fufa Fisheries Company Limited, Ningde, 352103, China.
| | - Ziping Zhang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
6
|
Lee SG, Huang M, Obholzer ND, Sun S, Li W, Petrillo M, Dai P, Zhou Y, Cotanche DA, Megason SG, Li H, Chen ZY. Myc and Fgf Are Required for Zebrafish Neuromast Hair Cell Regeneration. PLoS One 2016; 11:e0157768. [PMID: 27351484 PMCID: PMC4924856 DOI: 10.1371/journal.pone.0157768] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 06/03/2016] [Indexed: 01/05/2023] Open
Abstract
Unlike mammals, the non-mammalian vertebrate inner ear can regenerate the sensory cells, hair cells, either spontaneously or through induction after hair cell loss, leading to hearing recovery. The mechanisms underlying the regeneration are poorly understood. By microarray analysis on a chick model, we show that chick hair cell regeneration involves the activation of proliferation genes and downregulation of differentiation genes. Both MYC and FGF are activated in chick hair cell regeneration. Using a zebrafish lateral line neuromast hair cell regeneration model, we show that the specific inhibition of Myc or Fgf suppresses hair cell regeneration, demonstrating that both pathways are essential to the process. Rapid upregulation of Myc and delayed Fgf activation during regeneration suggest a role of Myc in proliferation and Fgf in differentiation. The dorsal-ventral pattern of fgfr1a in the neuromasts overlaps with the distribution of hair cell precursors. By laser ablation, we show that the fgfr1a-positive supporting cells are likely the hair cell precursors that directly give rise to new hair cells; whereas the anterior-posterior fgfr1a-negative supporting cells have heightened proliferation capacity, likely to serve as more primitive progenitor cells to replenish lost precursors after hair cell loss. Thus fgfr1a is likely to mark compartmentalized supporting cell subtypes with different capacities in renewal proliferation and hair cell regeneration. Manipulation of c-MYC and FGF pathways could be explored for mammalian hair cell regeneration.
Collapse
Affiliation(s)
- Sang Goo Lee
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Mingqian Huang
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Nikolaus D. Obholzer
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shan Sun
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
- Department of Otorhinolaryngology, Shanghai Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenyan Li
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
- Department of Otorhinolaryngology, Shanghai Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Marco Petrillo
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Pu Dai
- Department of Otolaryngology, Chinese PLA General Hospital, Beijing, China
| | - Yi Zhou
- Stem Cell Program and Division of Pediatric Hematology/Oncology, Children’s Hospital Boston and Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Douglas A. Cotanche
- Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Sean G. Megason
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Huawei Li
- Department of Otorhinolaryngology, Shanghai Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail: (ZYC); (HL)
| | - Zheng-Yi Chen
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
- * E-mail: (ZYC); (HL)
| |
Collapse
|
7
|
van Marion DM, Domanska UM, Timmer-Bosscha H, Walenkamp AM. Studying cancer metastasis: Existing models, challenges and future perspectives. Crit Rev Oncol Hematol 2016; 97:107-17. [DOI: 10.1016/j.critrevonc.2015.08.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/05/2015] [Indexed: 02/03/2023] Open
|
8
|
Brzuzan P, Kramer C, Łakomiak A, Jakimiuk E, Florczyk M, Woźny M. c-myc in whitefish (Coregonus lavaretus): structure, expression, and insights into possible posttranscriptional regulatory mechanism. FISH PHYSIOLOGY AND BIOCHEMISTRY 2015; 41:1155-1171. [PMID: 25995172 DOI: 10.1007/s10695-015-0077-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/15/2015] [Indexed: 06/04/2023]
Abstract
c-myc has a crucial function in growth control, differentiation, and apoptosis of vertebrate cells. Despite the important role of c-myc in mediating the biological effects, studies of c-myc gene expression and factors that control it in organisms other than mammals, such as fish, have been rare. In the current study, we asked whether c-myc mRNA of whitefish, a feasible organism for pollution monitoring in aquatic systems and a model in toxicological research, contains activity sites for regulatory motifs in its 5'- and 3'-UTRs, similar to those found in mammals. We were particularly interested in whether miRNA-34, a known negative regulator of c-myc's in mammals, is able to regulate c-myc in fish. To answer these questions, we determined the mRNA sequence of whitefish c-myc and inferred the structure of the protein that it codes for. We found that the active sites of mRNA and structures of the inferred c-myc protein are similar to those found in mammals and other fish. Remarkably, levels of c-myc mRNA expression were very high in ovaries compared to other tissues of whitefish, thus corroborating previous data in fish. Using bioinformatic searches on c-myc 3'-UTR, we confirmed the presence of two miRNA-34a (miR-34a) response elements. Luciferase reporter assay showed that activity of reporters containing either the miR response elements or entire c-myc 3'-UTR was significantly reduced (p < 0.001) by ectopic expression of miR-34a. Therefore, we further investigated possible involvement of miR-34a in c-myc gene silencing by profiling the expression of both genes in livers of whitefish treated for 8, 24, 48 h with MC-LR, a potent c-myc inducer in mammals. Although the difference was only significant at p = 0.08, the expression of c-myc mRNA in challenged whitefish after 24 h of the treatment was notably higher than that in livers of control fish. Concurrently, we noticed slight but significant up-regulation of miR-34a after 24 and 48 h of the challenge (p < 0.05); however, we found no significant correlation of the c-myc mRNA levels and miR-34a expression. Together, these results suggest that miR-34a might regulate c-myc gene expression in whitefish liver; however, their involvement in MC-LR hepatotoxicity should be clarified in future studies.
Collapse
Affiliation(s)
- P Brzuzan
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, ul. Słoneczna 45G, 10-709, Olsztyn, Poland.
| | - C Kramer
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, ul. Słoneczna 45G, 10-709, Olsztyn, Poland
| | - A Łakomiak
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, ul. Słoneczna 45G, 10-709, Olsztyn, Poland
| | - E Jakimiuk
- Department of Veterinary Prevention and Feed Hygiene, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, ul. Oczapowskiego 13, 10-950, Olsztyn, Poland
| | - M Florczyk
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, ul. Słoneczna 45G, 10-709, Olsztyn, Poland
| | - M Woźny
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, ul. Słoneczna 45G, 10-709, Olsztyn, Poland
| |
Collapse
|
9
|
Kotkamp K, Kur E, Wendik B, Polok BK, Ben-Dor S, Onichtchouk D, Driever W. Pou5f1/Oct4 promotes cell survival via direct activation of mych expression during zebrafish gastrulation. PLoS One 2014; 9:e92356. [PMID: 24643012 PMCID: PMC3958507 DOI: 10.1371/journal.pone.0092356] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 02/17/2014] [Indexed: 01/29/2023] Open
Abstract
Myc proteins control cell proliferation, cell cycle progression, and apoptosis, and play important roles in cancer as well in establishment of pluripotency. Here we investigated the control of myc gene expression by the Pou5f1/Oct4 pluripotency factor in the early zebrafish embryo. We analyzed the expression of all known zebrafish Myc family members, myca, mycb, mych, mycl1a, mycl1b, and mycn, by whole mount in situ hybridization during blastula and gastrula stages in wildtype and maternal plus zygotic pou5f1 mutant (MZspg) embryos, as well as by quantitative PCR and in time series microarray data. We found that the broad blastula and gastrula stage mych expression, as well as late gastrula stage mycl1b expression, both depend on Pou5f1 activity. We analyzed ChIP-Seq data and found that both Pou5f1 and Sox2 bind to mych and mycl1b control regions. The regulation of mych by Pou5f1 appears to be direct transcriptional activation, as overexpression of a Pou5f1 activator fusion protein in MZspg embryos induced strong mych expression even when translation of zygotically expressed mRNAs was suppressed. We further showed that MZspg embryos develop enhanced apoptosis already during early gastrula stages, when apoptosis was not be detected in wildtype embryos. However, Mych knockdown alone did not induce early apoptosis, suggesting potentially redundant action of several early expressed myc genes, or combination of several pathways affected in MZspg. Experimental mych overexpression in MZspg embryos did significantly, but not completely suppress the apoptosis phenotype. Similarly, p53 knockdown only partially suppressed apoptosis in MZspg gastrula embryos. However, combined knockdown of p53 and overexpression of Mych completely rescued the MZspg apoptosis phenotype. These results reveal that Mych has anti-apoptotic activity in the early zebrafish embryo, and that p53-dependent and Myc pathways are likely to act in parallel to control apoptosis at these stages.
Collapse
Affiliation(s)
- Kay Kotkamp
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Esther Kur
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Björn Wendik
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Bożena K. Polok
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Shifra Ben-Dor
- Biological Services, Weizmann Institute of Science, Rehovot, Israel
| | - Daria Onichtchouk
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
- BIOSS - Centre for Biological Signalling Studies, Freiburg, Germany
| | - Wolfgang Driever
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
- BIOSS - Centre for Biological Signalling Studies, Freiburg, Germany
- * E-mail:
| |
Collapse
|
10
|
Kopecky B, Fritzsch B. The myc road to hearing restoration. Cells 2012; 1:667-98. [PMID: 24710525 PMCID: PMC3901154 DOI: 10.3390/cells1040667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 08/12/2012] [Accepted: 09/14/2012] [Indexed: 01/01/2023] Open
Abstract
Current treatments for hearing loss, the most common neurosensory disorder, do not restore perfect hearing. Regeneration of lost organ of Corti hair cells through forced cell cycle re-entry of supporting cells or through manipulation of stem cells, both avenues towards a permanent cure, require a more complete understanding of normal inner ear development, specifically the balance of proliferation and differentiation required to form and to maintain hair cells. Direct successful alterations to the cell cycle result in cell death whereas regulation of upstream genes is insufficient to permanently alter cell cycle dynamics. The Myc gene family is uniquely situated to synergize upstream pathways into downstream cell cycle control. There are three Mycs that are embedded within the Myc/Max/Mad network to regulate proliferation. The function of the two ear expressed Mycs, N-Myc and L-Myc were unknown less than two years ago and their therapeutic potentials remain speculative. In this review, we discuss the roles the Mycs play in the body and what led us to choose them to be our candidate gene for inner ear therapies. We will summarize the recently published work describing the early and late effects of N-Myc and L-Myc on hair cell formation and maintenance. Lastly, we detail the translational significance of our findings and what future work must be performed to make the ultimate hearing aid: the regeneration of the organ of Corti.
Collapse
Affiliation(s)
- Benjamin Kopecky
- Department of Biology, 143 Biology Building, University of Iowa, Iowa City, IA 52242, USA.
| | - Bernd Fritzsch
- Department of Biology, 143 Biology Building, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
11
|
Menescal LA, Schmidt C, Liedtke D, Schartl M. Liver hyperplasia after tamoxifen induction of Myc in a transgenic medaka model. Dis Model Mech 2012; 5:492-502. [PMID: 22422827 PMCID: PMC3380712 DOI: 10.1242/dmm.008730] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Myc is a global transcriptional regulator and one of the most frequently overexpressed oncoproteins in human tumors. It is well established that activation of Myc leads to enhanced cell proliferation but can also lead to increased apoptosis. The use of animal models expressing deregulated levels of Myc has helped to both elucidate its function in normal cells and give insight into how Myc initiates and maintains tumorigenesis. Analyses of the medaka (Oryzias latipes) genome uncovered the unexpected presence of two Myc gene copies in this teleost species. Comparison of these Myc versions to other vertebrate species revealed that one gene, myc17, differs by the loss of some conserved regulatory protein motifs present in all other known Myc genes. To investigate how such differences might affect the basic biological functions of Myc, we generated a tamoxifen-inducible in vivo model utilizing a natural, fish-specific Myc gene. Using this model we show that, when activated, Myc17 leads to increased proliferation and to apoptosis in a dose-dependent manner, similar to human Myc. We have also shown that long-term Myc17 activation triggers liver hyperplasia in adult fish, allowing this newly established transgenic medaka model to be used to study the transition from hyperplasia to liver cancer and to identify Myc-induced tumorigenesis modifiers.
Collapse
Affiliation(s)
- Luciana A Menescal
- Physiological Chemistry I, University of Würzburg, Biozentrum, Am Hubland, D-97074 Würzburg, Germany
| | | | | | | |
Collapse
|
12
|
Marandel L, Labbe C, Bobe J, Le Bail PY. Evolutionary history of c-myc in teleosts and characterization of the duplicated c-myca genes in goldfish embryos. Mol Reprod Dev 2011; 79:85-96. [PMID: 22213278 DOI: 10.1002/mrd.22004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 11/04/2011] [Indexed: 01/13/2023]
Abstract
c-Myc plays an important role during embryogenesis in mammals, but little is known about its function during embryonic development in teleosts. In addition, the evolutionary history of c-myc gene in teleosts remains unclear, and depending on the species, a variable number of gene duplicates exist in teleosts. To gain new insight into c-myc genes in teleosts, the present study was designed to clarify the evolutionary history of c-myc gene(s) in teleosts and to subsequently characterize DNA methylation and early embryonic expression patterns in a cyprinid fish. Our results show that a duplication of c-myc gene occurred before or around the teleost radiation, as a result of the teleost-specific whole genome duplication giving rise to c-myca and c-mycb in teleosts and was followed by a loss of the c-mycb gene in the Gasterosteiforms and Tetraodontiforms. Our data also demonstrate that both c-myc genes previously identified in carp and goldfish are co-orthologs of the zebrafish c-myca. These results indicate the presence of additional c-myca duplication in Cyprininae. We were able to identify differences between the expression patterns of the two goldfish c-myca genes in oocytes and early embryos. These differences suggest a partial sub-functionalization of c-myca genes after duplication. Despite differences in transcription patterns, both of the c-myca genes displayed similar DNA methylation patterns during early development and in gametes. Together, our results clarify the evolutionary history of the c-myc gene in teleosts and provide new insight into the involvement of c-myc in early embryonic development in cyprinids.
Collapse
Affiliation(s)
- Lucie Marandel
- INRA, UR1037 SCRIBE, IFR140, Biogenouest, Rennes, France.
| | | | | | | |
Collapse
|
13
|
Stephens WZ, Senecal M, Nguyen M, Piotrowski T. Loss of adenomatous polyposis coli (apc) results in an expanded ciliary marginal zone in the zebrafish eye. Dev Dyn 2010; 239:2066-77. [PMID: 20549742 DOI: 10.1002/dvdy.22325] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The distal region of neural retina (ciliary marginal zone [CMZ]) contains stem cells that produce non-neural and neuronal progenitors. We provide a detailed gene expression analysis of the eyes of apc mutant zebrafish where the Wnt/beta-catenin pathway is constitutively active. Wnt/beta-catenin signaling leads to an expansion of the CMZ accompanied by a central shift of the retinal identity gene sox2 and the proneural gene atoh7. This suggests an important role for peripheral Wnt/beta-catenin signaling in regulating the expression and localization of neurogenic genes in the central retina. Retinal identity genes rx1 and vsx2, as well as meis1 and pax6a act upstream of Wnt/beta-catenin pathway activation. Peripheral cells that likely contain stem cells can be identified by the expression of follistatin, otx1, and axin2 and the lack of expression of myca and cyclinD1. Our results introduce the zebrafish apc mutation as a new model to study signaling pathways regulating the CMZ.
Collapse
Affiliation(s)
- W Zac Stephens
- Department of Neurobiology and Anatomy, University of Utah Medical School, 20N Medical Drive, Salt Lake City, UT 84132, USA
| | | | | | | |
Collapse
|
14
|
Feijóo CG, Saldias MP, De la Paz JF, Gómez-Skarmeta JL, Allende ML. Formation of posterior cranial placode derivatives requires the Iroquois transcription factor irx4a. Mol Cell Neurosci 2009; 40:328-37. [DOI: 10.1016/j.mcn.2008.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 09/02/2008] [Accepted: 11/17/2008] [Indexed: 01/29/2023] Open
|
15
|
Sanosaka T, Namihira M, Asano H, Kohyama J, Aisaki K, Igarashi K, Kanno J, Nakashima K. Identification of genes that restrict astrocyte differentiation of midgestational neural precursor cells. Neuroscience 2008; 155:780-8. [PMID: 18640244 DOI: 10.1016/j.neuroscience.2008.06.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 05/22/2008] [Accepted: 06/13/2008] [Indexed: 01/21/2023]
Abstract
During development of the mammalian CNS, neurons and glial cells (astrocytes and oligodendrocytes) are generated from common neural precursor cells (NPCs). However, neurogenesis precedes gliogenesis, which normally commences at later stages of fetal telencephalic development. Astrocyte differentiation of mouse NPCs at embryonic day (E) 14.5 (relatively late gestation) is induced by activation of the transcription factor signal transducer and activator of transcription (STAT) 3, whereas at E11.5 (mid-gestation) NPCs do not differentiate into astrocytes even when stimulated by STAT3-activating cytokines such as leukemia inhibitory factor (LIF). This can be explained in part by the fact that astrocyte-specific gene promoters are highly methylated in NPCs at E11.5, but other mechanisms are also likely to play a role. We therefore sought to identify genes involved in the inhibition of astrocyte differentiation of NPCs at midgestation. We first examined gene expression profiles in E11.5 and E14.5 NPCs, using Affymetrix GeneChip analysis, applying the Percellome method to normalize gene expression level. We then conducted in situ hybridization analysis for selected genes found to be highly expressed in NPCs at midgestation. Among these genes, we found that N-myc and high mobility group AT-hook 2 (Hmga2) were highly expressed in the E11.5 but not the E14.5 ventricular zone of mouse brain, where NPCs reside. Transduction of N-myc and Hmga2 by retroviruses into E14.5 NPCs, which normally differentiate into astrocytes in response to LIF, resulted in suppression of astrocyte differentiation. However, sustained expression of N-myc and Hmga2 in E11.5 NPCs failed to maintain the hypermethylated status of an astrocyte-specific gene promoter. Taken together, our data suggest that astrocyte differentiation of NPCs is regulated not only by DNA methylation but also by genes whose expression is controlled spatio-temporally during brain development.
Collapse
Affiliation(s)
- T Sanosaka
- Laboratory of Molecular Neuroscience, Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma, Nara 630-0101, Japan
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Hong SK, Tsang M, Dawid IB. The mych gene is required for neural crest survival during zebrafish development. PLoS One 2008; 3:e2029. [PMID: 18446220 PMCID: PMC2323570 DOI: 10.1371/journal.pone.0002029] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2007] [Accepted: 03/14/2008] [Indexed: 12/01/2022] Open
Abstract
Background Among Myc family genes, c-Myc is known to have a role in neural crest specification in Xenopus and in craniofacial development in the mouse. There is no information on the function of other Myc genes in neural crest development, or about any developmental role of zebrafish Myc genes. Principal Findings We isolated the zebrafish mych (myc homologue) gene. Knockdown of mych leads to severe defects in craniofacial development and in certain other tissues including the eye. These phenotypes appear to be caused by cell death in the neural crest and in the eye field in the anterior brain. Significance Mych is a novel factor required for neural crest cell survival in zebrafish.
Collapse
Affiliation(s)
- Sung-Kook Hong
- Laboratory of Molecular Genetics, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | | | |
Collapse
|
17
|
The c-myc coding DNA sequences of cyprinids (Teleostei: Cypriniformes): Implications for phylogeny. CHINESE SCIENCE BULLETIN-CHINESE 2007. [DOI: 10.1007/s11434-007-0216-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
18
|
Salierno J, Snyder N, Murphy A, Poli M, Hall S, Baden D, Kane A. Harmful algal bloom toxins alter c-Fos protein expression in the brain of killifish, Fundulus heteroclitus. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2006; 78:350-7. [PMID: 16750577 PMCID: PMC2659846 DOI: 10.1016/j.aquatox.2006.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 04/21/2006] [Accepted: 04/21/2006] [Indexed: 05/07/2023]
Abstract
The immediate early gene c-fos, and its protein product c-Fos, are known to be induced in neurons of mammals and fish as a result of neuronal stimulation. The purpose of this study was to quantitatively examine CNS alterations in killifish, Fundulus heteroclitus, in relation to harmful algal bloom (HAB) toxin exposure. c-Fos expression was visualized using immunocytochemistry in the brains of killifish exposed to the excitatory neurotoxins domoic acid (DA) and brevetoxin (PbTx-2), and a paralytic neurotoxin, saxitoxin (STX), released from HABs. In addition, a simulated transport stress experiment was conducted to investigate effects of physical stress on c-Fos induction. Groups of fish were exposed to the different stress agents, brain sections were processed for c-Fos staining, and expression was quantified by brain region. Fish exposed to DA, STX, and transport stress displayed significant alterations in neuronal c-Fos expression when compared to control fish (p< or = 0.05). DA, PbTx-2, and transport stress increased c-Fos expression in the optic tecta regions of the brain, whereas STX significantly decreased expression. This is the first study to quantify c-Fos protein expression in fish exposed to HAB toxins. General alterations in brain activity, as well as knowledge of specific regions within the brain activated in association with HABs or other stressors, provides valuable insights into the neural control of fish behavior as well as sublethal effects of specific stressors in the CNS.
Collapse
Affiliation(s)
- J.D. Salierno
- Aquatic Pathobiology Center, Department of Epidemiology and Preventive Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - N.S. Snyder
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - A.Z. Murphy
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - M. Poli
- US Army Medical Research Institute of Infectious Diseases, Integrated Toxicology Division, Fort Detrick, USA
| | - S. Hall
- US Food and Drug Administration, Office of Seafood, Beltsville Research Facility, Laurel, MD 20708, USA
| | - D. Baden
- Center for Marine Science Research, University of North Carolina Wilmington, Wilmington, NC 28409, USA
| | - A.S. Kane
- Aquatic Pathobiology Center, Department of Epidemiology and Preventive Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Aquatic Pathobiology Center, Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD 20742, USA
- Corresponding author. Tel.: +1 301 314 6808. E-mail address: (A.S. Kane)
| |
Collapse
|
19
|
Abstract
The Myc transcription factor regulates fundamental processes in a cell's life: its growth, division, and survival. Myc is conserved throughout metazoan phyla, and its identification in the fruit fly, Drosophila melanogaster has led to new insights in Myc's physiological roles. In this review, we describe recent research on the biology of Myc and its family members in Drosophila, paying particular attention to its role in the control of growth during development.
Collapse
Affiliation(s)
| | - Laura A. Johnston
- Corresponding author. Tel.: +1 212 305 1688; fax: +1 212 305 1752. (L.A. Johnston)
| |
Collapse
|
20
|
Futami K, Zhang H, Okamoto N. Functional divergence of duplicated c-myc genes in a tetraploid fish, the common carp (Cyprinus carpio). Gene 2005; 363:61-6. [PMID: 16242865 DOI: 10.1016/j.gene.2005.06.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Accepted: 06/27/2005] [Indexed: 11/20/2022]
Abstract
The proto-oncogene c-myc is thought to be one of the most important genes in controlling cell proliferation. In a tetraploid fish, two c-myc genes (CAM1 and CAM2) were previously isolated from the common carp, Cyprinus carpio, and were shown to have different expression patterns in adult tissues. Here we found that CAM1 and CAM2 proteins had distinct properties in terms of their transcription regulation system, formation of the transcription activator complex Myc/Max, and transcriptional activation of the target gene. These results showed that the two carp c-Myc proteins have overlapping but distinct functions, suggesting that CAM1 and CAM2 are evolving to acquire different functions after an earlier tetraploidization event.
Collapse
Affiliation(s)
- Kunihiko Futami
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology, Konan 4, Minato-ku, Tokyo 108-8477, Japan
| | | | | |
Collapse
|
21
|
Berghmans S, Jette C, Langenau D, Hsu K, Stewart R, Look T, Kanki JP. Making waves in cancer research: new models in the zebrafish. Biotechniques 2005; 39:227-37. [PMID: 16116796 DOI: 10.2144/05392rv02] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The zebrafish (Danio rerio) has proven to be a powerful vertebrate model system for the genetic analysis of developmental pathways and is only beginning to be exploited as a model for human disease and clinical research. The attributes that have led to the emergence of the zebrafish as a preeminent embryological model, including its capacity for forward and reverse genetic analyses, provides a unique opportunity to uncover novel insights into the molecular genetics of cancer. Some of the advantages of the zebrafish animal model system include fecundity, with each female capable of laying 200-300 eggs per week, external fertilization that permits manipulation of embryos ex utero, and rapid development of optically clear embryos, which allows the direct observation of developing internal organs and tissues in vivo. The zebrafish is amenable to transgenic and both forward and reverse genetic strategies that can be used to identify or generate zebrafish models of different types of cancer and may also present significant advantages for the discovery of tumor suppressor genes that promote tumorigenesis when mutationally inactivated. Importantly, the transparency and accessibility of the zebrafish embryo allows the unprecedented direct analysis of pathologic processes in vivo, including neoplastic cell transformation and tumorigenic progression. Ultimately, high-throughput modifier screens based on zebrafish cancer models can lead to the identification of chemicals or genes involved in the suppression or prevention of the malignant phenotype. The identification of small molecules or gene products through such screens will serve as ideal entry points for novel drug development for the treatment of cancer. This review focuses on the current technology that takes advantage of the zebrafish model system to further our understanding of the genetic basis of cancer and its treatment.
Collapse
|
22
|
Yamaguchi M, Tonou-Fujimori N, Komori A, Maeda R, Nojima Y, Li H, Okamoto H, Masai I. Histone deacetylase 1 regulates retinal neurogenesis in zebrafish by suppressing Wnt and Notch signaling pathways. Development 2005; 132:3027-43. [PMID: 15944187 DOI: 10.1242/dev.01881] [Citation(s) in RCA: 174] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the developing vertebrate retina, progenitor cells initially proliferate but begin to produce postmitotic neurons when neuronal differentiation occurs. However, the mechanism that determines whether retinal progenitor cells continue to proliferate or exit from the cell cycle and differentiate is largely unknown. Here, we report that histone deacetylase 1 (Hdac1) is required for the switch from proliferation to differentiation in the zebrafish retina. We isolated a zebrafish mutant, ascending and descending (add), in which retinal cells fail to differentiate into neurons and glial cells but instead continue to proliferate. The cloning of the add gene revealed that it encodes Hdac1. Furthermore, the ratio of the number of differentiating cells to that of proliferating cells increases in proportion to Hdac activity, suggesting that Hdac proteins regulate a crucial step of retinal neurogenesis in zebrafish. Canonical Wnt signaling promotes the proliferation of retinal cells in zebrafish, and Notch signaling inhibits neuronal differentiation through the activation of a neurogenic inhibitor, Hairy/Enhancer-of-split (Hes). We found that both the Wnt and Notch/Hes pathways are activated in the add mutant retina. The cell-cycle progression and the upregulation of Hes expression in the add mutant retina can be inhibited by the blockade of Wnt and Notch signaling, respectively. These data suggest that Hdac1 antagonizes these pathways to promote cell-cycle exit and the subsequent neurogenesis in zebrafish retina. Taken together, these data suggest that Hdac1 functions as a dual switch that suppresses both cell-cycle progression and inhibition of neurogenesis in the zebrafish retina.
Collapse
Affiliation(s)
- Masahiro Yamaguchi
- Masai Initiative Research Unit, RIKEN (The Institute of Physical and Chemical Research
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Loeb-Hennard C, Kremmer E, Bally-Cuif L. Prominent transcription of zebrafish N-myc (nmyc1) in tectal and retinal growth zones during embryonic and early larval development. Gene Expr Patterns 2005; 5:341-7. [PMID: 15661639 DOI: 10.1016/j.modgep.2004.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2004] [Revised: 10/15/2004] [Accepted: 10/15/2004] [Indexed: 10/26/2022]
Abstract
Because of its oncogenic capacity and expression restricted to embryonic and newborn tissues, the N-myc proto-oncogene is suggested to play a key role in vertebrate organogenesis as well as in the control of cell proliferation and differentiation. To further approach the developmental function of N-myc, we cloned full-length zebrafish N-myc (nmyc1) and analyzed its expression in the embryo and early larva. nmyc1 transcription is initiated at the mid-blastula stage. At somitogenesis stages, its expression was detected in the retina, midbrain, posterior hindbrain and presumptive spinal cord. nmyc1 was also transcribed in the endoderm and its derivatives as well as in branchial arches. At later developmental stages, posterior neural expression of nmyc1 was switched off, but expression remained intense in the brain, mainly in the optic tectum, cerebellar plate and dorsal rhombomere 2. Comparison of nmyc1 transcription with proliferation zones using a M phase mitotic marker revealed that nmyc1 expression is specifically associated with mitosis in the optic tectum and the retina. This result contrasts with previous studies in other vertebrates where N-myc expression can persist in differentiating cells.
Collapse
Affiliation(s)
- Christine Loeb-Hennard
- GSF-Research Center for Environment and Health, Institute of Molecular Immunology, Marchioninistrasse 25, Munich 81377, Germany.
| | | | | |
Collapse
|
24
|
Yimlamai D, Konnikova L, Moss LG, Jay DG. The zebrafish down syndrome cell adhesion molecule is involved in cell movement during embryogenesis. Dev Biol 2005; 279:44-57. [PMID: 15708557 DOI: 10.1016/j.ydbio.2004.12.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2004] [Revised: 11/30/2004] [Accepted: 12/01/2004] [Indexed: 10/26/2022]
Abstract
The Down syndrome cell adhesion molecule (Dscam) is a protein overexpressed in the brains of Down syndrome patients and implicated in mental retardation. Dscam is involved in axon guidance and branching in Drosophila, but cellular roles in vertebrates have yet to be elucidated. To understand its role in vertebrate development, we cloned the zebrafish homolog of Dscam and showed that it shares high amino acid identity and structure with the mammalian homologs. Zebrafish dscam is highly expressed in developing neurons, similar to what has been described in Drosophila and mouse. When dscam expression is diminished by morpholino injection, embryos display few neurons and their axons do not enter stereotyped pathways. Zebrafish dscam is also present at early embryonic stages including blastulation and gastrulation. Its loss results in early morphogenetic defects. dscam knockdown results in impaired cell movement during epiboly as well as in subsequent stages. We propose that migrating cells utilize dscam to remodel the developing embryo.
Collapse
Affiliation(s)
- Dean Yimlamai
- Cellular and Molecular Physiology, Tufts University School of Medicine, 136 Harrison Avenue, M and V 709, Boston, MA 02111, USA
| | | | | | | |
Collapse
|
25
|
Onnebo SMN, Yoong SHS, Ward AC. Harnessing zebrafish for the study of white blood cell development and its perturbation. Exp Hematol 2004; 32:789-96. [PMID: 15345279 DOI: 10.1016/j.exphem.2004.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Considerable progress has been made in understanding the molecular basis of normal white blood cell development and its perturbation in disease through the use of clinical studies and traditional animal and cell line models. Despite this, however, many questions are still being answered and white blood cell disorders, including leukemia and lymphoma, remain a significant health problem. The zebrafish (Danio rerio) has emerged as a powerful alternative vertebrate model for the study of development and disease. We review the recent application of zebrafish to the study of white blood cell development and its disruption, particularly leukemogenesis. Such studies have highlighted the overall conservation of these processes throughout vertebrates, and establish zebrafish as a useful experimental model. This organism is now poised to make an important contribution to our understanding of the underlying genetic control of white blood cell development and its disruption, as well as the identification of new therapeutic agents.
Collapse
Affiliation(s)
- Sara M N Onnebo
- Centre for Cellular & Molecular Biology, School of Biological & Chemical Sciences, Deakin University, Burwood, Victoria, Australia
| | | | | |
Collapse
|
26
|
Abstract
Fish have a long history of use in cancer toxicology studies, because they develop neoplasms that are histologically similar to human cancers. Because of considerable progress in zebrafish genetics and genomics over the past few years, the zebrafish system has provided many useful tools for studying basic biological processes. These tools include forward genetic screens, transgenic models, specific gene disruptions and small-molecule screens. By combining carcinogenesis assays, genetic analyses and small-molecule screening techniques, the zebrafish is emerging as a powerful system for identifying novel cancer genes and for cancer drug discovery.
Collapse
Affiliation(s)
- Howard M Stern
- Howard Hughes Medical Institute, Children's Hospital of Boston, 300 Longwood Avenue, Enders 761, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
27
|
Dorsky RI, Itoh M, Moon RT, Chitnis A. Two tcf3 genes cooperate to pattern the zebrafish brain. Development 2003; 130:1937-47. [PMID: 12642497 DOI: 10.1242/dev.00402] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Caudalizing factors operate in the context of Wnt/beta-catenin signaling to induce gene expression in discrete compartments along the rostral-caudal axis of the developing vertebrate nervous system. In zebrafish, basal repression of caudal genes is achieved through the function of Headless (Hdl), a Tcf3 homolog. In this study, we show that a second Tcf3 homolog, Tcf3b, limits caudalization caused by loss of Hdl function and although this Lef/Tcf family member can rescue hdl mutants, Lef1 cannot. Wnts can antagonize repression mediated by Tcf3 and this derepression is dependent on a Tcf3 beta-catenin binding domain. Systematic changes in gene expression caused by reduced Tcf3 function help predict the shape of a caudalizing activity gradient that defines compartments along the rostral-caudal axis. In addition, Tcf3b has a second and unique role in the morphogenesis of rhombomere boundaries, indicating that it controls multiple aspects of brain development.
Collapse
Affiliation(s)
- Richard I Dorsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA.
| | | | | | | |
Collapse
|
28
|
Spitsbergen JM, Kent ML. The state of the art of the zebrafish model for toxicology and toxicologic pathology research--advantages and current limitations. Toxicol Pathol 2003; 31 Suppl:62-87. [PMID: 12597434 PMCID: PMC1909756 DOI: 10.1080/01926230390174959] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The zebrafish (Danio rerio) is now the pre-eminent vertebrate model system for clarification of the roles of specific genes and signaling pathways in development. The zebrafish genome will be completely sequenced within the next 1-2 years. Together with the substantial historical database regarding basic developmental biology, toxicology, and gene transfer, the rich foundation of molecular genetic and genomic data makes zebrafish a powerful model system for clarifying mechanisms in toxicity. In contrast to the highly advanced knowledge base on molecular developmental genetics in zebrafish, our database regarding infectious and noninfectious diseases and pathologic lesions in zebrafish lags far behind the information available on most other domestic mammalian and avian species, particularly rodents. Currently, minimal data are available regarding spontaneous neoplasm rates or spontaneous aging lesions in any of the commonly used wild-type or mutant lines of zebrafish. Therefore, to fully utilize the potential of zebrafish as an animal model for understanding human development, disease, and toxicology we must greatly advance our knowledge on zebrafish diseases and pathology.
Collapse
Affiliation(s)
- Jan M Spitsbergen
- Department of Environmental and Molecular Toxicology and Marine/Freshwater Biomedical Sciences Center, Oregon State University, Corvallis, Oregon 97333, USA.
| | | |
Collapse
|
29
|
Abstract
The fish kidney provides a unique model for investigating renal injury, repair, and development. Like mammalian kidneys, fish kidneys have the remarkable ability to repair injured nephrons, designated renal regeneration. This response is marked by a recovery from acute renal failure by replacing the injured cells with new epithelial cells, restoring tubule integrity. In addition, fish have the ability to respond to renal injury by de novo nephron neogenesis. This response occurs in multiple fish species including goldfish, zebrafish, catfish, trout, tilapia, and the aglomerular toadfish. New nephrons develop in the weeks after the initial injury. This nephrogenic response can be induced in adult fish, providing a more abundant source of developing renal tissue compared with fetal mammalian kidneys. Investigating the roles played by different parts of the nephron during development and repair can be facilitated using fish models with differing renal anatomy, such as aglomerular fish. The fish nephron neogenesis model may also help to identify novel genes involved in nephrogenesis, information that could eventually be used to develop alternative renal replacement therapies.
Collapse
Affiliation(s)
- R Reimschuessel
- Office of Research, Center for Veterinary Medicine, US Food and Drug Administration, Laurel, Maryland, USA
| |
Collapse
|
30
|
Futami K, Komiya T, Zhang H, Okamoto N. Differential expression of max and two types of c- myc genes in a tetraploid fish, the common carp ( Cyprinus carpio ). Gene 2001; 269:113-9. [PMID: 11376943 DOI: 10.1016/s0378-1119(01)00453-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We cloned the full-length cDNA of max gene from the common carp (Cyprinus carpio). The cDNA clone of carp max consists of 1209 bp and contained an ATG-initiated ORF consisting of 156 aa. The carp MAX share 76.7-93.8% aa identity with those of human, mouse, rat, chicken, Xenopus and zebrafish, respectively. The 15 bp alternative splicing was observed in the loop region of helix-loop-helix and is not previously described in mammalian max sequences. Transcripts of max gene were observed in all of the tissues of carp investigated in this study. The highest expression was found in the ovary, and the transcripts in hepatopancreas and heart were low. Two carp c-myc genes (CAM1 and CAM2) showed differential expression pattern. The expression of max was concomitant with CAM2 expression, but not with CAM1. It has been reported that MYC/MAX heterodimer as a regulator of gene expression has been maintained throughout vertebrate evolution, and the expression of c-myc has been concomitant with max expression. In addition, according to phylogenetic analysis, CAM1 is evolving faster than CAM2 after gene duplication. Therefore, this result suggests that CAM1 may evolve to obtain a new function different from c-myc.
Collapse
Affiliation(s)
- K Futami
- Department of Aquatic Biosciences, Tokyo University of Fisheries, Konan 4, Minato-ku, 108-8477, Tokyo, Japan
| | | | | | | |
Collapse
|
31
|
Murphy M, Harte T, McInerney J, Smith TJ. Molecular cloning of an Atlantic salmon nucleoside diphosphate kinase cDNA and its pattern of expression during embryogenesis. Gene 2000; 257:139-48. [PMID: 11054576 DOI: 10.1016/s0378-1119(00)00374-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
To gain insight into the process of development in Atlantic salmon (Salmo salar), we sought to identify genes that were differentially expressed at gastrulation. A polymerase chain reaction-based differential screening strategy allowed for the isolation of an Atlantic salmon nucleoside diphosphate kinase cDNA (nm23). Structural characterisation showed a high degree of homology with a large number of previously isolated nucleoside diphosphate kinases (NM23s), both prokaryote and eukaryote, though it represents the first teleost nucleoside diphosphate kinase identified. Highest similarities were found with the type 1 and type 2 NM23 isoforms of mammals. Phylogenetic analysis indicates that the duplication event that gave rise to these isoforms occurred after the splitting of tetrapods and fish, suggesting that the salmon NM23 represents a more ancestral isoform. The position of the salmon sequence on the phylogenetic tree indicates that the salmon genome is expected to have at least three copies of genes from the nm23 gene family. Northern blot analysis showed a single transcript of approximately 0.7 kb in both embryonic and adult tissues. Examination of the temporal pattern of expression of salmon nucleoside diphosphate kinase during embryonic development revealed that this gene is first expressed at the time of gastrulation. Nucleoside diphosphate kinases are thought to have a vital role in regulatory processes such as signal transduction, proliferation and differentiation. Taken together, these results suggest that nucleoside diphosphate kinases have an important role to play in early embryogenic development in vertebrates.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Blotting, Northern
- Cloning, Molecular
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- DNA, Complementary/isolation & purification
- Embryo, Nonmammalian/enzymology
- Embryonic Development
- Gene Expression Regulation, Developmental
- Gene Expression Regulation, Enzymologic
- Molecular Sequence Data
- Nucleoside-Diphosphate Kinase/genetics
- Phylogeny
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Salmo salar/embryology
- Salmo salar/genetics
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Tissue Distribution
Collapse
Affiliation(s)
- M Murphy
- BioResearch Ireland, National Diagnostics Centre, National University of Ireland, Galway, Ireland
| | | | | | | |
Collapse
|
32
|
Abstract
Apoptosis has been well established as a vital biological phenomenon that is important in the maintenance of cellular homeostasis. Three major protooncogene families and their encoded proteins function as mediators of apoptosis in various cell types and are the subject of this chapter. Protooncogenic proteins such as c-Myc/Max, c-Fos/c-Jun, and Bcl-2/Bax utilize a synergetic effect to enhance their roles in the pro- or antiapoptotic action. These family members activate and repress the expression of their target genes, control cell cycle progression, and execute programmed cell death. Repression or overproduction of these protooncogenic proteins induces apoptosis, which may vary as a result of either cell type specificity or the nature of the apoptotic stimuli. The proapoptotic and antiapoptotic proteins exert their effects in the membrane of cellular organelles. Here they generate cell-type-specific signals that activate the caspase family of proteases and their regulators for the execution of apoptosis.
Collapse
Affiliation(s)
- C S Teng
- Department of Anatomy, Physiological Sciences, and Radiology, North Carolina State University, Raleigh 27606, USA
| |
Collapse
|
33
|
Malynn BA, de Alboran IM, O'Hagan RC, Bronson R, Davidson L, DePinho RA, Alt FW. N-myc can functionally replace c-myc in murine development, cellular growth, and differentiation. Genes Dev 2000; 14:1390-9. [PMID: 10837031 PMCID: PMC316670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Members of the myc family of cellular oncogenes have been implicated as transcriptional regulators in pathways that govern cellular proliferation and death. In addition, N-myc and c-myc are essential for completion of murine embryonic development. However, the basis for the evolutionary conservation of myc gene family has remained unclear. To elucidate this issue, we have generated mice in which the endogenous c-myc coding sequences have been replaced with N-myc coding sequences. Strikingly, mice homozygous for this replacement mutation can survive into adulthood and reproduce. Moreover, when expressed from the c-myc locus, N-myc is similarly regulated and functionally complementary to c-myc in the context of various cellular growth and differentiation processes. Therefore, the myc gene family must have evolved, to a large extent, to facilitate differential patterns of expression.
Collapse
Affiliation(s)
- B A Malynn
- The Center for Blood Research, Boston, Massachusetts 02115 USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Stenkamp DL, Frey RA, Prabhudesai SN, Raymond PA. Function for Hedgehog genes in zebrafish retinal development. Dev Biol 2000; 220:238-52. [PMID: 10753513 DOI: 10.1006/dbio.2000.9629] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The hedgehog (hh) genes encode secreted signaling proteins that have important developmental functions in vertebrates and invertebrates. In Drosophila, expression of hh coordinates retinal development by propagating a wave of photoreceptor differentiation across the eye primordium. Here we report that two vertebrate hh genes, sonic hedgehog (shh) and tiggy-winkle hedgehog (twhh), may perform similar functions in the developing zebrafish. Both shh and twhh are expressed in the embryonic zebrafish retinal pigmented epithelium (RPE), initially in a discrete ventral patch which then expands outward in advance of an expanding wave of photoreceptor recruitment in the subjacent neural retina. A gene encoding a receptor for the hedgehog protein, ptc-2, is expressed by retinal neuroepithelial cells. Injection of a cocktail of antisense (alphashh/alphatwhh) oligonucleotides reduces expression of both hh genes in the RPE and slows or arrests the progression of rod and cone photoreceptor differentiation. Zebrafish strains known to have mutations in Hh signaling pathway genes similarly exhibit retardation of photoreceptor differentiation. We propose that hedgehog genes may play a role in propagating photoreceptor differentiation across the developing eye of the zebrafish.
Collapse
Affiliation(s)
- D L Stenkamp
- Department of Biological Sciences, University of Idaho, Moscow, Idaho 83844-3051, USA
| | | | | | | |
Collapse
|
35
|
Futami K, Komiya T, Zhang H, Okamoto N. Determination of heterogeneous transcription start points of two c-myc genes from the common carp (Cyprinus carpio). Gene 2000; 245:43-7. [PMID: 10713443 DOI: 10.1016/s0378-1119(00)00044-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We determined the heterogeneous transcription start points (tsp) of two c-myc genes from the common carp (Cyprinus carpio), tetraploid teleost, by the oligo-capping method and showed the existence of the first exon. This is the first report on the existence of the first exons of the fish c-myc gene. Transcription of the two carp c-myc genes started from at least four sites in CAM1, locating from -752 to -381bp upstream of the translation start site, and from 12 sites in CAM2, locating from -586 to -413bp upstream respectively. The first introns of CAM1 and CAM2 were deduced to be 335 and 356bp, respectively. They shared 86.9% nt identity, lower than those of the second exons (94.1%), and third exons (92.3%), which suggest that the first exons evolved faster. No nt identities were found between the c-myc first exons of carp and other vertebrates. The putative promoter regions in CAM1 and CAM2 contained no obvious TATA or CCAAT boxes in the expected positions.
Collapse
Affiliation(s)
- K Futami
- Department of Aquatic Biosciences, Tokyo University of Fisheries, Konan 4, Minato-ku, Tokyo, Japan
| | | | | | | |
Collapse
|
36
|
Tsai JN, Lee CH, Jeng H, Chi WK, Chang WC. Differential expression of glycogen synthase kinase 3 genes during zebrafish embryogenesis. Mech Dev 2000; 91:387-91. [PMID: 10704871 DOI: 10.1016/s0925-4773(99)00300-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Glycogen synthase kinase 3 (GSK-3) belongs to a highly conserved family of protein serine/threonine kinase whose members in high eukaryotes are involved in hormonal regulation, nuclear signaling, and cell fate determination. We have identified two zebrafish homologues related to mammalian GSK-3, ZGSK-3alpha and ZGSK-3beta. ZGSK-3alpha was expressed uniformly from cleavage onward, and later was found in many but not all tissues, especially in the central nervous system, spinal cord, somites and pronephric ducts. ZGSK-3beta was also transcribed maternally but the transcripts were not uniformly distributed during early cleavage stage. Most signals were concentrated in the inner part of the blastomeres. From midblastula stage onward, the ZGSK-3beta transcripts remained confined to inner parts of the deep cell layer. During shield stage, both epiblast and hypoblast expressed the transcripts. After late gastrulation, the signals were detected ubiquitously. During segmentation, prominent ZGSK-3beta signal was detected in head portion of the neural system. In the trunk, the expression was maintained in the neural tube and paraxial mesoderm and then became prominent in adaxial cells, followed by expression at the posterior region of somites. In pharyngula period ZGSK-3beta transcripts were distributed in similar regions as those of ZGSK-3alpha, namely, neural tissues of the head portion, spinal cord and somites.
Collapse
Affiliation(s)
- J N Tsai
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
37
|
Liu YW, Lo LJ, Chan WK. Temporal expression and T3 induction of thyroid hormone receptors alpha1 and beta1 during early embryonic and larval development in zebrafish, Danio rerio. Mol Cell Endocrinol 2000; 159:187-95. [PMID: 10687864 DOI: 10.1016/s0303-7207(99)00193-8] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The effects of thyroid hormones on metabolism and development are mediated by thyroid hormone receptors (TRs). We report the cloning and characterization of a TR beta1 cDNA from zebrafish. Southern blot analysis revealed that there is a single genomic locus for the TR beta gene, while the TR alpha gene potentially has two loci. Multiple TR alpha and TR beta transcripts were detected in adult tissues. Using a semiquantitative RT-PCR assay, zygotic expression of TR alpha1 and TR beta1 were shown to occur before the midblastula transition stage. In transiently transfected HeLa cells, TR alpha1 displayed constitutive transactivation in the absence of ligands, which was slightly enhanced by triiodothyronine (T3). The transactivating activity of TR beta1 was strictly ligand-dependent and repressed in the absence of T3. Finally, the T3 induction of TR alpha1 and TR beta1 mRNAs was demonstrated in zebrafish embryos and larvae. The auto-induction of TR alpha1 and TR beta1 may serve a regulatory role during the embryonic and larval development of zebrafish.
Collapse
Affiliation(s)
- Y W Liu
- Laboratory of Fish Biology, Institute of Molecular Agrobiology, National University of Singapore, Singapore
| | | | | |
Collapse
|
38
|
Shen-Li H, O'Hagan RC, Hou H, Horner JW, Lee HW, DePinho RA. Essential role for Max in early embryonic growth and development. Genes Dev 2000; 14:17-22. [PMID: 10640271 PMCID: PMC316346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Loss of Max function in the mouse resulted in generalized developmental arrest of both embryonic and extraembryonic tissues at early postimplantation (approximately E5.5-6.5), coincident with loss or dilution of maternal Max stores in the expanding embryo in vivo and in blastocyst outgrowths in vitro. Developmentally arrested embryos were reduced in size and exhibited widespread cytological degeneration and feeble BrdU incorporation. Max and, by extension, the Myc superfamily, serve essential roles in early mammalian development and a maternal reservoir of Max exists in sufficient amount to sustain Myc superfamily function through preimplantation stages of development.
Collapse
Affiliation(s)
- H Shen-Li
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York 10461 USA
| | | | | | | | | | | |
Collapse
|
39
|
Lüscher B, Larsson LG. The basic region/helix-loop-helix/leucine zipper domain of Myc proto-oncoproteins: function and regulation. Oncogene 1999; 18:2955-66. [PMID: 10378692 DOI: 10.1038/sj.onc.1202750] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A large body of evidence has been accumulated that demonstrates dominant effects of Myc proto-oncoproteins on different aspects of cellular growth. Myc is one of the few proteins that is sufficient to drive resting cells into the cell cycle and promote DNA synthesis. In line with this finding is that the constitutive expression of Myc in cells blocks their differentiation. These growth stimulating properties are most likely responsible for Myc's ability to initiate and promote tumor formation. Interestingly Myc can also sensitize cells to apoptosis, suggesting that this protein is part of a life-and-death switch. Molecularly Myc functions as a transcriptional regulator that needs to heterodimerize with Max to exert the biological activities described above and to regulate gene transcription. Myc and Max are just two members of a growing family of proteins referred to as the Myc/Max/Mad network. A hallmark of these proteins is that they possess a C-terminal basic region/helix-loop-helix/leucine zipper domain (bHLHZip). The bHLHZip domain specifies dimerization within the network and determines sequence specific DNA binding. Importantly this domain together with the N-terminal transactivation domain is essential for Myc biology. Here we have summarized the structural, functional, and regulatory aspects of the bHLHZip domain of Myc proteins.
Collapse
Affiliation(s)
- B Lüscher
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Germany.
| | | |
Collapse
|
40
|
Wang X, Peters MA, Utama FE, Wang Y, Taparowsky EJ. The Adrenomedullin gene is a target for negative regulation by the Myc transcription complex. Mol Endocrinol 1999; 13:254-67. [PMID: 9973255 DOI: 10.1210/mend.13.2.0240] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The Myc family of transcription factors plays a central role in vertebrate growth and development although relatively few genetic targets of the Myc transcription complex have been identified. In this study, we used mRNA differential display to investigate gene expression changes induced by the overexpression of the MC29 v-Myc oncoprotein in C3H10T1/2 mouse fibroblasts. We identified the transcript of the adrenomedullin gene (AM) as an mRNA that is specifically down-regulated in v-Myc overexpressing C3H10T1/2 cell lines as well as in a Rat 1a cell line inducible for c-Myc. Nucleotide sequence analysis of the mouse AM promoter reveals the presence of consensus CAAT and TATA boxes as well as an initiator element (INR) with significant sequence similarity to the INR responsible for Myc-mediated repression of the adenovirus major late promoter (AdMLP). Reporter gene assays confirm that the region of the AM promoter containing the INR is the target of Myc-mediated repression. Exogenous application of AM peptide to quiescent C3H10T1/2 cultures does not stimulate growth, and constitutive expression of AM mRNA in C3H10T1/2 cells correlates with a reduced potential of the cells to be cotransformed by v-Myc and oncogenic Ras p21. Additional studies showing that AM mRNA is underrepresented in C3H10T1/2 cell lines stably transformed by Ras p21 or adenovirus E1A suggest that AM gene expression is incompatible with deregulated growth in this cell line. We propose a model in which the repression of AM gene expression by Myc is important to the role of this oncoprotein as a potentiator of cellular transformation in C3H10T1/2 and perhaps other cell lines.
Collapse
Affiliation(s)
- X Wang
- Department of Biological Sciences, Purdue University West Lafayette, Indiana 47907-1392, USA
| | | | | | | | | |
Collapse
|
41
|
Kelly GM, Reversade B. Characterization of a cDNA encoding a novel band 4.1-like protein in zebrafish. Biochem Cell Biol 1997. [DOI: 10.1139/o97-078] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Membrane skeleton protein 4.1 and other members of a family of proteins that link the cytoskeleton to the plasma membrane may play an integral role in cell communication during development. The polymerase chain reaction and degenerate oligodeoxynucleotide primers to consensus sequences in the putative membrane-binding domain of the protein 4.1 superfamily were used to isolate cDNAs encoding members of the zebrafish protein 4.1 family. Zebrafish stage- and tissue-specific first strand cDNA was used in the PCR. After the reaction, amplicons of the predicted size were sequenced to confirm their relationship to the protein 4.1 superfamily. One cDNA, with a high degree of similarity to a mouse novel band 4.1-like cDNA, was used to probe a zebrafish adult brain library. A 2.4-kb cDNA was isolated and found to encode a 619 amino acid polypeptide homologous to mouse novel band 4.1-like protein 4. Zebrafish nbl4 mRNA is maternally supplied and is expressed throughout embryogenesis. In adults, nbl4 is found in the ovary, eye, heart, and brain, but not in gut or skeletal muscle. When synthetic nbl4 mRNA is translated in vitro it binds calmodulin in a calcium-dependent manner. These data indicate that zebrafish nbl4 is a maternal transcript owing to its presence before the midblastula transition, and it is present later on in specific adult structures. The ability to bind calmodulin would suggest that the function of nbl4 protein may be potentially regulated via a calcium-calmodulin dependent mechanism.
Collapse
|
42
|
Carter CA, Ellington WW, Van Beneden RJ. Confocal laser scanning microscopy of oncogene localization in rainbow trout cell lines derived from normal and tumor tissue. Toxicol Pathol 1996; 24:339-45. [PMID: 8736390 DOI: 10.1177/019262339602400310] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We examined the localization and expression of the nuclear oncoprotein c-myc and the cytoplasmic membrane-associated oncoprotein c-ras in rainbow trout cell lines derived from both normal and tumor tissue in order to question whether c-myc and ras oncoprotein immunostaining was increased in cells derived from tumors compared to cells derived from normal tissue. Cell lines examined were derived from normal rainbow trout gonadal cells (RTG-2), a rainbow trout hepatoma (RTH-149), and a rainbow trout mesothelioma (RTM). Protein products of c-ras and c-myc were visualized in these 3 cell lines by employing fluorescein-labeled anti-mouse pan-ras and c-myc antibodies. The RTG-2 cells were used in this study as normal, control cells, and they exhibited little pan-ras and c-myc staining. The RTH-149 cell line (a tumorigenic cell line) exhibited positive pan-ras staining in regions of the membrane and cell cytoplasm. Localization of c-myc staining to perinuclear regions was punctate in RTH-149 cells. RTM cells (also a tumorigenic cell line) displayed a ras staining localization similar to the pattern seen in RTH-149 cells. RTM cells exhibit a diffuse perinuclear staining and, thus, display a more ubiquitous localization of c-myc than RTH-149 cells. Northern blot analysis indicated that c-myc expression was highest in RTM cells, whereas RTG-2 cells and RTH-149 cells expressed similar lower levels of c-myc expression. We were unable to detect significant ras expression in any of the cell lines by Northern blot analysis. In summary, the cell line derived from normal tissue, the RTG-2 cells, displayed little ras and c-myc immunostaining, whereas the cell lines derived from tumorigenic tissue, RTH and RTM cells, displayed increased immunostaining for c-myc and ras proteins.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Cell Line
- Fish Diseases/genetics
- Fish Diseases/metabolism
- Fish Diseases/pathology
- Genes, myc/genetics
- Genes, ras/genetics
- Gonads/cytology
- Immunohistochemistry
- Liver Neoplasms, Experimental/chemically induced
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Mesothelioma/metabolism
- Mesothelioma/pathology
- Mesothelioma/veterinary
- Microscopy, Confocal
- Neoplasms/metabolism
- Neoplasms/pathology
- Neoplasms/veterinary
- Oncorhynchus mykiss/genetics
- Oncorhynchus mykiss/metabolism
- Phosphorus Radioisotopes
- Proto-Oncogene Proteins c-myc/biosynthesis
- Proto-Oncogene Proteins c-myc/metabolism
- RNA, Messenger/biosynthesis
- RNA, Neoplasm/biosynthesis
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- C A Carter
- Laboratory of Experimental Pathology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | | | | |
Collapse
|
43
|
Henriksson M, Lüscher B. Proteins of the Myc network: essential regulators of cell growth and differentiation. Adv Cancer Res 1996; 68:109-82. [PMID: 8712067 DOI: 10.1016/s0065-230x(08)60353-x] [Citation(s) in RCA: 585] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- M Henriksson
- Institute for Molecular Biology, Hannover Medical School, Germany
| | | |
Collapse
|
44
|
Lemaitre JM, Buckle RS, Méchali M. c-Myc in the control of cell proliferation and embryonic development. Adv Cancer Res 1996; 70:95-144. [PMID: 8902055 DOI: 10.1016/s0065-230x(08)60873-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
45
|
Rebollo A, Gómez J, Martínez-A C. Lessons from immunological, biochemical, and molecular pathways of the activation mediated by IL-2 and IL-4. Adv Immunol 1996; 63:127-96. [PMID: 8787631 DOI: 10.1016/s0065-2776(08)60856-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- A Rebollo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Universidad Autónoma de Madrid, Spain
| | | | | |
Collapse
|
46
|
Panno JP, McKeown BA. Cloning and expression of a myc family member from the pituitary gland of the Rainbow trout, Oncorhynchus mykiss. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1264:7-11. [PMID: 7578259 DOI: 10.1016/0167-4781(95)00121-v] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A myc gene cloned from a Rainbow trout pituitary gland cDNA library is described. This clone (Tmyc2) shows extensive homology to Rainbow trout C-myc, which is expressed in the liver. Tmyc2 does not appear in the liver but is expressed in the pituitary gland (1.9 kb transcript), brain (2.0 kb transcript) and, at very low levels, in the heart (2.0 kb transcript). Tmyc2 contains three highly modified areas within the coding sequence, one of which shows an extensive loss of acidic residues that is uncommon in C-myc family members and may be important in determining the function of Tmyc2 in the pituitary gland and brain.
Collapse
Affiliation(s)
- J P Panno
- Department of Biological Sciences, Simon Fraser University, Burnaby, B.C., Canada
| | | |
Collapse
|
47
|
Kelly GM, Erezyilmaz DF, Moon RT. Induction of a secondary embryonic axis in zebrafish occurs following the overexpression of beta-catenin. Mech Dev 1995; 53:261-73. [PMID: 8562427 DOI: 10.1016/0925-4773(95)00442-4] [Citation(s) in RCA: 106] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Formation of the vertebrate axis may involve a Wnt signaling cascade similar to the Drosophila wingless pathway. Zebrafish wnt8 is a candidate for involvement in axis specification insofar as it is expressed maternally and when overexpressed it can induce goosecoid, a transcription factor normally expressed in the embryonic shield. In this study we demonstrate that beta-catenin, a cadherin associated protein in the Wnt pathway, is expressed maternally in zebrafish and is widely distributed in the early embryo. Overexpressing beta-catenin in early zebrafish embryos induces goosecoid and ntl, ultimately leading to a duplication of a complete secondary axis. These data are consistent with the involvement of beta-catenin in a Wnt signaling pathway which is involved in mesoderm induction in zebrafish.
Collapse
Affiliation(s)
- G M Kelly
- Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle 98195-7370, USA
| | | | | |
Collapse
|
48
|
Abstract
The Wnts are a family of secreted glycoproteins involved in cell-cell signalling and pattern formation during development, although the extent to which various Wnts are functionally equivalent remains unclear. We have cloned zebrafish (Danio rerio) wnt4, characterized its expression, and compared its activity relative to other Wnts. The wnt4 transcript is first detected early in somitogenesis, in the dorsocaudal region of the forebrain, and then appears in the dorsal and lateral regions of the caudal hindbrain and neural keel. During somitogenesis, wnt4 appears in the floor plate, and this expression is absent in cyclops mutants, which lack floor plate. wnt4 is also expressed in the developing pronephros and gill slit. To characterize the biological activity of wnt4, synthetic zebrafish wnt4 mRNA was injected into embryos of zebrafish and Xenopus laevis. The phenotypic effects of misexpression in the zebrafish include cyclopia, misfolding in the brain, and an anteriorly forking notochord. Comparison of the phenotypes arising from misexpression of wnt4 and Xwnt-5A in both organisms suggests close parallels in the response to these Wnts. Our data suggest that wnt4, like Xwnt-5A, inhibits cell movements, and that these Wnts define a functional class distinct from the class which includes Wnt-1, Xwnt-3A and Xwnt-8.
Collapse
Affiliation(s)
- A R Ungar
- Department of Pharmacology, University of Washington School of Medicine, Seattle 98195, USA
| | | | | |
Collapse
|
49
|
Kelly GM, Greenstein P, Erezyilmaz DF, Moon RT. Zebrafish wnt8 and wnt8b share a common activity but are involved in distinct developmental pathways. Development 1995; 121:1787-99. [PMID: 7600994 DOI: 10.1242/dev.121.6.1787] [Citation(s) in RCA: 171] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The specification of the vertebrate body plan is dependent on numerous signaling molecules, including members of the Wnt family. We have identified two zebrafish wnt8 paralogs related to Xwnt-8B and Xwnt-8, respectively. A RT-PCR assay demonstrated that wnt8 is expressed maternally, with transcripts detected throughout embryogenesis, whereas wnt8b transcripts were first detected during late gastrulation. The wnt8 transcripts at 50% epiboly are spatially restricted to those cells at the blastoderm margin, overlying gsc-expressing cells in the axial hypoblast. During late gastrulation, wnt8 was no longer detected in the marginal cells at the dorsal midline and by mid-segmentation, transcripts were found in the presumptive tail bud. In contrast, wnt8b expression is spatially restricted to prospective neuroepithelium, and later to neural-specific structures. Overexpression of both wnts results in two major phenotypes: radialized embryos and embryos with anterior defects. These phenotypes were preceded by significant changes in the spatial expression patterns of gsc and ntl transcripts, reminiscent of activities of Xwnt-8 in Xenopus, and consistent with a role for wnt8 in the specification or patterning of mesoderm.
Collapse
Affiliation(s)
- G M Kelly
- Howard Hughes Medical Institute, Department of Pharmacology, Seattle, Washington, USA
| | | | | | | |
Collapse
|
50
|
Bosch TJ, Maslam S, Roberts BL. A polyclonal antibody against mammalian FOS can be used as a cytoplasmic neuronal activity marker in a teleost fish. J Neurosci Methods 1995; 58:173-9. [PMID: 7475225 DOI: 10.1016/0165-0270(94)00174-f] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A polyclonal antibody raised against a conserved region of a mammalian FOS sequence was tested for its use as an activity marker in the rainbow trout. The FOS-like expression in the trout is entirely cytoplasmic and appears in a Nissl-like pattern. The reaction is specifically induced by both orthodromic and antidromic electrical stimuli and during motor responses evoked by natural stimulation, although some positive neurons are found at locations that are not obviously related to the presented stimuli. Following spinal nerve stimulation, antidromically activated motoneurons were found to be positive in the ipsilateral spinal cord. Orthodromic driving of spinal moto- and interneurons by stimulation of the medial longitudinal fasciculus (MLF) in the hindbrain evoked FOS-like immunoreactivity throughout the motor column in the spinal cord, but not in regions lying caudal to a lesion of the MLF-axons. Evoking about 25 startle responses by natural auditory stimulation gives FOS-like immunoreactivity in the Mauthner cell, which initiates the response, whereas positive Mauthner cells were never observed in control fish. The stimulation protocols that were used strongly activated the stimulated cells and so the observed FOS-like immunoreactivity might be related to an increase protein synthesis needed to restore their depleted transmitter levels.
Collapse
Affiliation(s)
- T J Bosch
- Department of Experimental Zoology, University of Amsterdam, The Netherlands
| | | | | |
Collapse
|