Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Mol Cell ; 82(23): 4410-4427.e12, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36356583

ABSTRACT

Gene expression heterogeneity underlies cell states and contributes to developmental robustness. While heterogeneity can arise from stochastic transcriptional processes, the extent to which it is regulated is unclear. Here, we characterize the regulatory program underlying heterogeneity in murine embryonic stem cell (mESC) states. We identify differentially active and transcribed enhancers (DATEs) across states. DATEs regulate differentially expressed genes and are distinguished by co-binding of transcription factors Klf4 and Zfp281. In contrast to other factors that interact in a positive feedback network stabilizing mESC cell-type identity, Klf4 and Zfp281 drive opposing transcriptional and chromatin programs. Abrogation of factor binding to DATEs dampens variation in gene expression, and factor loss alters kinetics of switching between states. These results show antagonism between factors at enhancers results in gene expression heterogeneity and formation of cell states, with implications for the generation of diverse cell types during development.


Subject(s)
Embryonic Stem Cells , Transcription Factors , Animals , Mice , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Proc Natl Acad Sci U S A ; 119(19): e2107006119, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35512098

ABSTRACT

Cutaneous melanoma (CM) and uveal melanoma (UM) both originate from the melanocytic lineage but are primarily driven by distinct oncogenic drivers, BRAF/NRAS or GNAQ/GNA11, respectively. The melanocytic master transcriptional regulator, MITF, is essential for both CM development and maintenance, but its role in UM is largely unexplored. Here, we use zebrafish models to dissect the key UM oncogenic signaling events and establish the role of MITF in UM tumors. Using a melanocytic lineage expression system, we showed that patient-derived mutations of GNAQ (GNAQQ209L) or its upstream CYSLTR2 receptor (CYSLTR2L129Q) both drive UM when combined with a cooperating mutation, tp53M214K/M214K. The tumor-initiating potential of the major GNAQ/11 effector pathways, YAP, and phospholipase C-ß (PLCß)­ERK was also investigated in this system and thus showed that while activated YAP (YAPAA) induced UM with high potency, the patient-derived PLCß4 mutation (PLCB4D630Y) very rarely yielded UM tumors in the tp53M214K/M214K context. Remarkably, mitfa deficiency was profoundly UM promoting, dramatically accelerating the onset and progression of tumors induced by Tg(mitfa:GNAQQ209L);tp53M214K/M214K or Tg(mitfa:CYSLTR2L129Q);tp53M214K/M214K. Moreover, mitfa loss was sufficient to cooperate with GNAQQ209L to drive tp53­wild type UM development and allowed Tg(mitfa:PLCB4D630Y);tp53M214K/M214K melanocyte lineage cells to readily form tumors. Notably, all of the mitfa−/− UM tumors, including those arising in Tg(mitfa:PLCB4D630Y);tp53M214K/M214K;mitfa−/− zebrafish, displayed nuclear YAP while lacking hyperactive ERK indicative of PLCß signaling. Collectively, these data show that YAP signaling is the major mediator of UM and that MITF acts as a bona fide tumor suppressor in UM in direct opposition to its essential role in CM.


Subject(s)
Melanoma , Skin Neoplasms , Uveal Neoplasms , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Uveal Neoplasms/therapy , Melanoma, Cutaneous Malignant
3.
Genesis ; 62(1): e23568, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37946671

ABSTRACT

Epithelial-mesenchymal transition (EMT) and primary ciliogenesis are two cell-biological programs that are essential for development of multicellular organisms and whose abnormal regulation results in many diseases (i.e., developmental anomalies and cancers). Emerging studies suggest an intricate interplay between these two processes. Here, we discuss physiological and pathological contexts in which their interconnections promote normal development or disease progression. We describe underlying molecular mechanisms of the interplay and EMT/ciliary signaling axes that influence EMT-related processes (i.e., stemness, motility and invasion). Understanding the molecular and cellular mechanisms of the relationship between EMT and primary ciliogenesis may provide new insights in the etiology of diseases related to EMT and cilia dysfunction.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms , Humans , Epithelial-Mesenchymal Transition/physiology , Signal Transduction , Cilia
4.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Article in English | MEDLINE | ID: mdl-34408017

ABSTRACT

Epigenetic regulators play key roles in cancer and are increasingly being targeted for treatment. However, for many, little is known about mechanisms of resistance to the inhibition of these regulators. We have generated a model of resistance to inhibitors of protein arginine methyltransferase 5 (PRMT5). This study was conducted in KrasG12D;Tp53-null lung adenocarcinoma (LUAD) cell lines. Resistance to PRMT5 inhibitors (PRMT5i) arose rapidly, and barcoding experiments showed that this resulted from a drug-induced transcriptional state switch, not selection of a preexisting population. This resistant state is both stable and conserved across variants arising from distinct LUAD lines. Moreover, it brought with it vulnerabilities to other chemotherapeutics, especially the taxane paclitaxel. This paclitaxel sensitivity depended on the presence of stathmin 2 (STMN2), a microtubule regulator that is specifically expressed in the resistant state. Remarkably, STMN2 was also essential for resistance to PRMT5 inhibition. Thus, a single gene is required for both acquisition of resistance to PRMT5i and collateral sensitivity to paclitaxel in our LUAD cells. Accordingly, the combination of PRMT5i and paclitaxel yielded potent and synergistic killing of the murine LUAD cells. Importantly, the synergy between PRMT5i and paclitaxel also extended to human cancer cell lines. Finally, analysis of The Cancer Genome Atlas patient data showed that high STMN2 levels correlate with complete regression of tumors in response to taxane treatment. Collectively, this study reveals a recurring mechanism of PRMT5i resistance in LUAD and identifies collateral sensitivities that have potential clinical relevance.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Paclitaxel/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Drug Synergism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mutation , Stathmin/genetics , Stathmin/metabolism
5.
Genes Dev ; 30(1): 18-33, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26679841

ABSTRACT

Melanoma can switch between proliferative and invasive states, which have identifying gene expression signatures that correlate with good and poor prognosis, respectively. However, the mechanisms controlling these signatures are poorly understood. In this study, we identify BMI1 as a key determinant of melanoma metastasis by which its overexpression enhanced and its deletion impaired dissemination. Remarkably, in this tumor type, BMI1 had no effect on proliferation or primary tumor growth but enhanced every step of the metastatic cascade. Consistent with the broad spectrum of effects, BMI1 activated widespread gene expression changes, which are characteristic of melanoma progression and also chemoresistance. Accordingly, we showed that up-regulation or down-regulation of BMI1 induced resistance or sensitivity to BRAF inhibitor treatment and that induction of noncanonical Wnt by BMI1 is required for this resistance. Finally, we showed that our BMI1-induced gene signature encompasses all of the hallmarks of the previously described melanoma invasive signature. Moreover, our signature is predictive of poor prognosis in human melanoma and is able to identify primary tumors that are likely to become metastatic. These data yield key insights into melanoma biology and establish BMI1 as a compelling drug target whose inhibition would suppress both metastasis and chemoresistance of melanoma.


Subject(s)
Drug Resistance, Neoplasm/genetics , Melanoma/genetics , Melanoma/physiopathology , Polycomb Repressive Complex 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/diagnosis , Melanoma/drug therapy , Mice , Neoplasm Invasiveness/genetics , Polycomb Repressive Complex 1/genetics , Prognosis , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms , Melanoma, Cutaneous Malignant
6.
Proc Natl Acad Sci U S A ; 117(14): 8001-8012, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32193336

ABSTRACT

The cyclin-dependent kinase 5 (CDK5), originally described as a neuronal-specific kinase, is also frequently activated in human cancers. Using conditional CDK5 knockout mice and a mouse model of highly metastatic melanoma, we found that CDK5 is dispensable for the growth of primary tumors. However, we observed that ablation of CDK5 completely abrogated the metastasis, revealing that CDK5 is essential for the metastatic spread. In mouse and human melanoma cells CDK5 promotes cell invasiveness by directly phosphorylating an intermediate filament protein, vimentin, thereby inhibiting assembly of vimentin filaments. Chemical inhibition of CDK5 blocks the metastatic spread of patient-derived melanomas in patient-derived xenograft (PDX) mouse models. Hence, inhibition of CDK5 might represent a very potent therapeutic strategy to impede the metastatic dissemination of malignant cells.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Melanoma, Experimental/pathology , Melanoma/pathology , Skin Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Female , Gene Dosage , Humans , Male , Melanoma/drug therapy , Melanoma/genetics , Melanoma/mortality , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/genetics , Prognosis , Skin/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Vimentin/metabolism , Xenograft Model Antitumor Assays
7.
Genes Dev ; 29(17): 1875-89, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26314710

ABSTRACT

The retinoblastoma tumor suppressor (pRb) protein associates with chromatin and regulates gene expression. Numerous studies have identified Rb-dependent RNA signatures, but the proteomic effects of Rb loss are largely unexplored. We acutely ablated Rb in adult mice and conducted a quantitative analysis of RNA and proteomic changes in the colon and lungs, where Rb(KO) was sufficient or insufficient to induce ectopic proliferation, respectively. As expected, Rb(KO) caused similar increases in classic pRb/E2F-regulated transcripts in both tissues, but, unexpectedly, their protein products increased only in the colon, consistent with its increased proliferative index. Thus, these protein changes induced by Rb loss are coupled with proliferation but uncoupled from transcription. The proteomic changes in common between Rb(KO) tissues showed a striking decrease in proteins with mitochondrial functions. Accordingly, RB1 inactivation in human cells decreased both mitochondrial mass and oxidative phosphorylation (OXPHOS) function. RB(KO) cells showed decreased mitochondrial respiratory capacity and the accumulation of hypopolarized mitochondria. Additionally, RB/Rb loss altered mitochondrial pyruvate oxidation from (13)C-glucose through the TCA cycle in mouse tissues and cultured cells. Consequently, RB(KO) cells have an enhanced sensitivity to mitochondrial stress conditions. In summary, proteomic analyses provide a new perspective on Rb/RB1 mutation, highlighting the importance of pRb for mitochondrial function and suggesting vulnerabilities for treatment.


Subject(s)
Mitochondria/metabolism , Oxidative Phosphorylation , Retinoblastoma Protein/genetics , Animals , Cells, Cultured , Colon/physiopathology , Gene Expression Regulation , Gene Knockout Techniques , Humans , Lung/physiopathology , Mice , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Proteomics , Retinoblastoma Protein/metabolism , Stress, Physiological/genetics , Transcriptome
8.
Genes Dev ; 27(9): 1003-15, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23618872

ABSTRACT

The retinoblastoma protein gene RB-1 is mutated in one-third of human tumors. Its protein product, pRB (retinoblastoma protein), functions as a transcriptional coregulator in many fundamental cellular processes. Here, we report a nonnuclear role for pRB in apoptosis induction via pRB's direct participation in mitochondrial apoptosis. We uncovered this activity by finding that pRB potentiated TNFα-induced apoptosis even when translation was blocked. This proapoptotic function was highly BAX-dependent, suggesting a role in mitochondrial apoptosis, and accordingly, a fraction of endogenous pRB constitutively associated with mitochondria. Remarkably, we found that recombinant pRB was sufficient to trigger the BAX-dependent permeabilization of mitochondria or liposomes in vitro. Moreover, pRB interacted with BAX in vivo and could directly bind and conformationally activate BAX in vitro. Finally, by targeting pRB specifically to mitochondria, we generated a mutant that lacked pRB's classic nuclear roles. This mito-tagged pRB retained the ability to promote apoptosis in response to TNFα and also additional apoptotic stimuli. Most importantly, induced expression of mito-tagged pRB in Rb(-/-);p53(-/-) tumors was sufficient to block further tumor development. Together, these data establish a nontranscriptional role for pRB in direct activation of BAX and mitochondrial apoptosis in response to diverse stimuli, which is profoundly tumor-suppressive.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation, Neoplastic , Mitochondria/metabolism , Retinoblastoma Protein/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cytochromes c/metabolism , Humans , Mice , Mice, Nude , Mice, SCID , Mitochondria/genetics , Protein Binding , Retinoblastoma Protein/genetics , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/metabolism
9.
Proc Natl Acad Sci U S A ; 114(49): E10532-E10539, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29158396

ABSTRACT

Tissue regeneration relies on adult stem cells (SCs) that possess the ability to self-renew and produce differentiating progeny. In an analogous manner, the development of certain carcinomas depends on a small subset of tumor cells, called "tumor-initiating cells" (TICs), with SC-like properties. Mammary SCs (MaSCs) reside in the basal compartment of the mammary epithelium, and their neoplastic counterparts, mammary TICs (MaTICs), are thought to serve as the TICs for the claudin-low subtype of breast cancer. MaSCs and MaTICs both use epithelial-mesenchymal transition (EMT) programs to acquire SC properties, but the mechanism(s) connecting EMT programs to stemness remain unclear. Here we show that this depends on primary cilia, which are nonmotile, cell-surface structures that serve as platforms for receiving cues and enable activation of various signaling pathways. We show that MaSC and MaTIC EMT programs induce primary cilia formation and Hedgehog (Hh) signaling, which has previously been implicated in both MaSC and MaTIC function. Moreover, ablation of these primary cilia is sufficient to repress Hh signaling, the stemness of MaSCs, and the tumor-forming potential of MaTICs. Together, our findings establish primary ciliogenesis and consequent Hh signaling as a key mechanism by which MaSC and MaTIC EMT programs promote stemness and thereby support mammary tissue outgrowth and tumors of basal origin.


Subject(s)
Cilia/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Animals , Cell Line, Tumor , Cilia/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, SCID , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Occludin/genetics , Occludin/metabolism , Signal Transduction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Vimentin/genetics , Vimentin/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
10.
Mol Carcinog ; 57(11): 1640-1650, 2018 11.
Article in English | MEDLINE | ID: mdl-30084175

ABSTRACT

Altered cell polarity and migration are hallmarks of cancer and metastases. Here we show that inactivation of the retinoblastoma gene (Rb) tumor suppressor causes defects in tissue closure that reflect the inability of Rb null epithelial cells to efficiently migrate and polarize. These defects occur independently of pRB's anti-proliferative role and instead correlate with upregulation of RhoA signaling and mislocalization of apical-basal polarity proteins. Notably, concomitant inactivation of tp53 specifically overrides the motility defect, and not the aberrant polarity, thereby uncovering previously unappreciated mechanisms by which Rb and tp53 mutations cooperate to promote cancer development and metastases.


Subject(s)
Cell Movement/genetics , Cell Polarity/genetics , Epithelial Cells/metabolism , Retinoblastoma Protein/genetics , Tumor Suppressor Proteins/genetics , Acute-Phase Proteins/metabolism , Animals , Gene Silencing , Humans , Mice , Mutation , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
11.
Am J Med Genet A ; 176(1): 92-98, 2018 01.
Article in English | MEDLINE | ID: mdl-29130579

ABSTRACT

The primary cilium is a key organelle in numerous physiological and developmental processes. Genetic defects in the formation of this non-motile structure, in its maintenance and function, underlie a wide array of ciliopathies in human, including craniofacial, brain and heart malformations, and retinal and hearing defects. We used exome sequencing to study the molecular basis of disease in an 11-year-old female patient who suffered from growth retardation, global developmental delay with absent speech acquisition, agenesis of corpus callosum and paucity of white matter, sensorineural deafness, retinitis pigmentosa, vertebral anomalies, patent ductus arteriosus, and facial dysmorphism reminiscent of STAR syndrome, a suspected ciliopathy. A homozygous variant, c.870_871del, was identified in the CDK10 gene, predicted to cause a frameshift, p.Trp291Alafs*18, in the cyclin-dependent kinase 10 protein. CDK10 mRNAs were detected in patient cells and do not seem to undergo non-sense mediated decay. CDK10 is the binding partner of Cyclin M (CycM) and CDK10/CycM protein kinase regulates ciliogenesis and primary cilium elongation. Notably, CycM gene is mutated in patients with STAR syndrome. Following incubation, the patient cells appeared less elongated and more densely populated than the control cells suggesting that the CDK10 mutation affects the cytoskeleton. Upon starvation and staining with acetylated-tubulin, γ-tubulin, and Arl13b, the patient cells exhibited fewer and shorter cilia than control cells. These findings underscore the importance of CDK10 for the regulation of ciliogenesis. CDK10 defect is likely associated with a new form of ciliopathy phenotype; additional patients may further validate this association.


Subject(s)
Agenesis of Corpus Callosum/genetics , Cyclin-Dependent Kinases , Deafness/genetics , Genetic Association Studies , Homozygote , Mutation , Retinal Dysplasia/genetics , Agenesis of Corpus Callosum/diagnosis , Alleles , Brain/abnormalities , Brain/diagnostic imaging , Child , DNA Mutational Analysis , Deafness/diagnosis , Exome , Facies , Female , Gene Expression , Humans , Pedigree , Phenotype , RNA, Messenger/genetics , Retinal Dysplasia/diagnosis
12.
Genes Dev ; 24(2): 183-94, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20080954

ABSTRACT

Eukaryotes have numerous checkpoint pathways to protect genome fidelity during normal cell division and in response to DNA damage. Through a screen for G2/M checkpoint regulators in zebrafish, we identified ticrr (for TopBP1-interacting, checkpoint, and replication regulator), a previously uncharacterized gene that is required to prevent mitotic entry after treatment with ionizing radiation. Ticrr deficiency is embryonic-lethal in the absence of exogenous DNA damage because it is essential for normal cell cycle progression. Specifically, the loss of ticrr impairs DNA replication and disrupts the S/M checkpoint, leading to premature mitotic entry and mitotic catastrophe. We show that the human TICRR ortholog associates with TopBP1, a known checkpoint protein and a core component of the DNA replication preinitiation complex (pre-IC), and that the TICRR-TopBP1 interaction is stable without chromatin and requires BRCT motifs essential for TopBP1's replication and checkpoint functions. Most importantly, we find that ticrr deficiency disrupts chromatin binding of pre-IC, but not prereplication complex, components. Taken together, our data show that TICRR acts in association with TopBP1 and plays an essential role in pre-IC formation. It remains to be determined whether Ticrr represents the vertebrate ortholog of the yeast pre-IC component Sld3, or a hitherto unknown metazoan replication and checkpoint regulator.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Replication/genetics , Genes, cdc/physiology , Mitosis/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Animals , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian , Humans , Mutation/genetics , Phenotype , Zebrafish/genetics
13.
Nature ; 466(7310): 1110-4, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20686481

ABSTRACT

Mutation of the retinoblastoma gene (RB1) tumour suppressor occurs in one-third of all human tumours and is particularly associated with retinoblastoma and osteosarcoma. Numerous functions have been ascribed to the product of the human RB1 gene, the retinoblastoma protein (pRb). The best known is pRb's ability to promote cell-cycle exit through inhibition of the E2F transcription factors and the transcriptional repression of genes encoding cell-cycle regulators. In addition, pRb has been shown in vitro to regulate several transcription factors that are master differentiation inducers. Depending on the differentiation factor and cellular context, pRb can either suppress or promote their transcriptional activity. For example, pRb binds to Runx2 and potentiates its ability to promote osteogenic differentiation in vitro. In contrast, pRb acts with E2F to suppress peroxisome proliferator-activated receptor gamma subunit (PPAR-gamma), the master activator of adipogenesis. Because osteoblasts and adipocytes can both arise from mesenchymal stem cells, these observations suggest that pRb might play a role in the choice between these two fates. However, so far, there is no evidence for this in vivo. Here we use mouse models to address this hypothesis in mesenchymal tissue development and tumorigenesis. Our data show that Rb status plays a key role in establishing fate choice between bone and brown adipose tissue in vivo.


Subject(s)
Adipose Tissue, Brown/cytology , Cell Differentiation , Cell Lineage , Osteoblasts/cytology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Lipoma/physiopathology , Mice , Mutation/genetics , PPAR gamma/metabolism , Sarcoma/physiopathology
14.
PLoS Genet ; 9(8): e1003734, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24009526

ABSTRACT

The identification of cancer drivers is a major goal of current cancer research. Finding driver genes within large chromosomal events is especially challenging because such alterations encompass many genes. Previously, we demonstrated that zebrafish malignant peripheral nerve sheath tumors (MPNSTs) are highly aneuploid, much like human tumors. In this study, we examined 147 zebrafish MPNSTs by massively parallel sequencing and identified both large and focal copy number alterations (CNAs). Given the low degree of conserved synteny between fish and mammals, we reasoned that comparative analyses of CNAs from fish versus human MPNSTs would enable elimination of a large proportion of passenger mutations, especially on large CNAs. We established a list of orthologous genes between human and zebrafish, which includes approximately two-thirds of human protein-coding genes. For the subset of these genes found in human MPNST CNAs, only one quarter of their orthologues were co-gained or co-lost in zebrafish, dramatically narrowing the list of candidate cancer drivers for both focal and large CNAs. We conclude that zebrafish-human comparative analysis represents a powerful, and broadly applicable, tool to enrich for evolutionarily conserved cancer drivers.


Subject(s)
Chromosome Aberrations , DNA Copy Number Variations/genetics , Genes, Neoplasm , Neurilemmoma/genetics , Aneuploidy , Animals , Gene Expression Regulation, Neoplastic , Genome, Human , Genomics , High-Throughput Nucleotide Sequencing , Humans , Neurilemmoma/pathology , Oligonucleotide Array Sequence Analysis , Zebrafish/genetics
15.
Cancer Cell ; 11(2): 175-89, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17292828

ABSTRACT

In response to DNA damage, eukaryotic cells activate ATM-Chk2 and/or ATR-Chk1 to arrest the cell cycle and initiate DNA repair. We show that, in the absence of p53, cells depend on a third cell-cycle checkpoint pathway involving p38MAPK/MK2 for cell-cycle arrest and survival after DNA damage. MK2 depletion in p53-deficient cells, but not in p53 wild-type cells, caused abrogation of the Cdc25A-mediated S phase checkpoint after cisplatin exposure and loss of the Cdc25B-mediated G2/M checkpoint following doxorubicin treatment, resulting in mitotic catastrophe and pronounced regression of murine tumors in vivo. We show that the Chk1 inhibitor UCN-01 also potently inhibits MK2, suggesting that its clinical efficacy results from the simultaneous disruption of two critical checkpoint pathways in p53-defective cells.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Protein Kinases/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Division/drug effects , Cell Division/radiation effects , Cell Survival , Cells, Cultured , Cisplatin/pharmacology , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Doxorubicin/pharmacology , G2 Phase/drug effects , G2 Phase/radiation effects , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Mice, Nude , Mitosis/drug effects , Mitosis/radiation effects , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Phosphorylation/drug effects , Phosphorylation/radiation effects , Protein Kinase C/antagonists & inhibitors , Protein Kinases/genetics , S Phase/drug effects , S Phase/radiation effects , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Ultraviolet Rays , cdc25 Phosphatases/metabolism
16.
Curr Opin Cell Biol ; 19(6): 649-57, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18032011

ABSTRACT

The E2F transcription factors are critical regulators of genes required for appropriate progression through the cell cycle, and in special circumstances they can also promote the expression of another class of genes that function in the apoptotic program. Since E2Fs can initiate both cell proliferation and cell death, it is not surprising that the pro-apoptotic capacity of these proteins is subject to complex regulation. Recent study has expanded our knowledge of the factors influencing E2F-induced apoptosis as well as downstream targets of E2F in this process.


Subject(s)
Apoptosis , Cell Proliferation , E2F Transcription Factors/physiology , Transcription, Genetic , Animals , Gene Expression Regulation , Humans
17.
Proc Natl Acad Sci U S A ; 107(39): 16940-5, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20837522

ABSTRACT

Aneuploidy is a hallmark of human cancers, but most mouse cancer models lack the extensive aneuploidy seen in many human tumors. The zebrafish is becoming an increasingly popular model for studying cancer. Here we report that malignant peripheral nerve sheath tumors (MPNSTs) that arise in zebrafish as a result of mutations in either ribosomal protein (rp) genes or in p53 are highly aneuploid. Karyotyping reveals that these tumors frequently harbor near-triploid numbers of chromosomes, and they vary in chromosome number from cell to cell within a single tumor. Using array comparative genomic hybridization, we found that, as in human cancers, certain fish chromosomes are preferentially overrepresented, whereas others are underrepresented in many MPNSTs. In addition, we obtained evidence for recurrent subchromosomal amplifications and deletions that may contain genes involved in cancer initiation or progression. These focal amplifications encompassed several genes whose amplification is observed in human tumors, including met, cyclinD2, slc45a3, and cdk6. One focal amplification included fgf6a. Increasing fgf signaling via a mutation that overexpresses fgf8 accelerated the onset of MPNSTs in fish bearing a mutation in p53, suggesting that fgf6a itself may be a driver of MPNSTs. Our results suggest that the zebrafish is a useful model in which to study aneuploidy in human cancer and in which to identify candidate genes that may act as drivers in fish and potentially also in human tumors.


Subject(s)
Aneuploidy , Disease Models, Animal , Nerve Sheath Neoplasms/genetics , Peripheral Nerves , Zebrafish/genetics , Animals , Fibroblast Growth Factors/genetics , Humans , Ribosomal Proteins/genetics , Sequence Deletion , Tumor Suppressor Protein p53/genetics , Zebrafish Proteins/genetics
18.
Pigment Cell Melanoma Res ; 36(5): 399-406, 2023 09.
Article in English | MEDLINE | ID: mdl-37132544

ABSTRACT

The epigenetic repressor BMI1 plays an integral role in promoting the self-renewal and proliferation of many adult stem cell populations, and also tumor types, primarily through silencing the Cdkn2a locus, which encodes the tumor suppressors p16Ink4a and p19Arf . However, in cutaneous melanoma, BMI1 drives epithelial-mesenchymal transition programs, and thus metastasis, while having little impact on proliferation or primary tumor growth. This raised questions about the requirement and role for BMI1 in melanocyte stem cell (McSC) biology. Here, we demonstrate that murine melanocyte-specific Bmi1 deletion causes premature hair greying and gradual loss of melanocyte lineage cells. Depilation enhances this hair greying defect, accelerating depletion of McSCs in early hair cycles, suggesting that BMI1 acts to protect McSCs against stress. RNA-seq of McSCs, harvested before onset of detectable phenotypic defects, revealed that Bmi1 deletion derepresses p16Ink4a and p19Arf , as observed in many other stem cell contexts. Additionally, BMI1 loss downregulated the glutathione S-transferase enzymes, Gsta1 and Gsta2, which can suppress oxidative stress. Accordingly, treatment with the antioxidant N-acetyl cysteine (NAC) partially rescued melanocyte expansion. Together, our data establish a critical function for BMI1 in McSC maintenance that reflects a partial role for suppression of oxidative stress, and likely transcriptional repression of Cdkn2a.


Subject(s)
Melanoma , Skin Neoplasms , Mice , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Melanoma/metabolism , Proto-Oncogene Proteins , Skin Neoplasms/metabolism , Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Pigmentation , Melanocytes/metabolism , Hair/metabolism
19.
Cancer Cell ; 2(6): 463-72, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12498715

ABSTRACT

The E2F transcription factors mediate the activation or repression of key cell cycle regulatory genes under the control of the retinoblastoma protein (pRB) tumor suppressor and its relatives, p107 and p130. Here we investigate how E2F4, the major "repressive" E2F, contributes to pRB's tumor-suppressive properties. Remarkably, E2F4 loss suppresses the development of both pituitary and thyroid tumors in Rb(+/-) mice. Importantly, E2F4 loss also suppresses the inappropriate gene expression and proliferation of pRB-deficient cells. Biochemical analyses suggest that this tumor suppression occurs via a novel mechanism: E2F4 loss allows p107 and p130 to regulate the pRB-specific, activator E2Fs. We also detect these novel E2F complexes in pRB-deficient cells, suggesting that they play a significant role in the regulation of tumorigenesis in vivo.


Subject(s)
DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Proteins , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Animals , Blotting, Western , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cyclin E/biosynthesis , DNA-Binding Proteins/metabolism , E2F4 Transcription Factor , Fibroblasts/metabolism , Mice , Mice, Mutant Strains , Mutation , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Pituitary Neoplasms/genetics , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/genetics , Transcription Factors/metabolism
20.
Proc Natl Acad Sci U S A ; 106(35): 14932-6, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19706423

ABSTRACT

The retinoblastoma tumor-suppressor protein, pRb, is a member of the pocket protein family that includes p107 and p130. These proteins have well-defined roles in regulating entry into and exit from the cell cycle and also have cell cycle-independent roles in facilitating differentiation. Here we investigate the overlap between pocket protein's function during embryonic development by using conditional mutant alleles to generate Rb;p107 double-mutant embryos (DKOs) that develop in the absence of placental defects. These DKOs die between e13.5 and e14.5, much earlier than either the conditional Rb or the germline p107 single mutants, which survive to birth or are largely viable, respectively. Analyses of the e13.5 DKOs shows that p107 mutation exacerbates the phenotypes resulting from pRb loss in the central nervous system and lens, but not in the peripheral nervous system. In addition, these embryos exhibit novel phenotypes, including increased proliferation of blood vessel endothelial cells, and heart defects, including double-outlet right ventricle (DORV). The DORV is caused, at least in part, by a defect in blood vessel endothelial cells and/or heart mesenchymal cells. These findings demonstrate novel, overlapping functions for pRb and p107 in numerous murine tissues.


Subject(s)
Embryo, Mammalian/metabolism , Heart Diseases/metabolism , Mutation , Retinoblastoma Protein/deficiency , Retinoblastoma-Like Protein p107/genetics , Animals , Apoptosis , Cell Proliferation , Central Nervous System/cytology , Central Nervous System/metabolism , Embryo, Mammalian/blood supply , Female , Gene Expression Regulation, Developmental , Heart Diseases/embryology , Heart Diseases/genetics , Heart Diseases/pathology , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Male , Mice , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL