Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Res ; 81(7): 1813-1826, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33495232

ABSTRACT

Small cell lung cancer (SCLC) is a pulmonary neuroendocrine cancer with very poor prognosis and limited effective therapeutic options. Most patients are diagnosed at advanced stages, and the exact reason for the aggressive and metastatic phenotype of SCLC is completely unknown. Despite a high tumor mutational burden, responses to immune checkpoint blockade are minimal in patients with SCLC. This may reflect defects in immune surveillance. Here we illustrate that evading natural killer (NK) surveillance contributes to SCLC aggressiveness and metastasis, primarily through loss of NK-cell recognition of these tumors by reduction of NK-activating ligands (NKG2DL). SCLC primary tumors expressed very low level of NKG2DL mRNA and SCLC lines express little to no surface NKG2DL at the protein level. Chromatin immunoprecipitation sequencing showed NKG2DL loci in SCLC are inaccessible compared with NSCLC, with few H3K27Ac signals. Restoring NKG2DL in preclinical models suppressed tumor growth and metastasis in an NK cell-dependent manner. Likewise, histone deacetylase inhibitor treatment induced NKG2DL expression and led to tumor suppression by inducing infiltration and activation of NK and T cells. Among all the common tumor types, SCLC and neuroblastoma were the lowest NKG2DL-expressing tumors, highlighting a lineage dependency of this phenotype. In conclusion, these data show that epigenetic silencing of NKG2DL results in a lack of stimulatory signals to engage and activate NK cells, highlighting the underlying immune avoidance of SCLC and neuroblastoma. SIGNIFICANCE: This study discovers in SCLC and neuroblastoma impairment of an inherent mechanism of recognition of tumor cells by innate immunity and proposes that this mechanism can be reactivated to promote immune surveillance.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Tumor Escape/physiology , Animals , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , HEK293 Cells , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Nude , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasm Metastasis , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/pathology , Tumor Escape/genetics
2.
Genes Dev ; 34(9-10): 621-636, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32241803

ABSTRACT

Peripheral somatosensory input is modulated in the dorsal spinal cord by a network of excitatory and inhibitory interneurons. PTF1A is a transcription factor essential in dorsal neural tube progenitors for specification of these inhibitory neurons. Thus, mechanisms regulating Ptf1a expression are key for generating neuronal circuits underlying somatosensory behaviors. Mutations targeted to distinct cis-regulatory elements for Ptf1a in mice, tested the in vivo contribution of each element individually and in combination. Mutations in an autoregulatory enhancer resulted in reduced levels of PTF1A, and reduced numbers of specific dorsal spinal cord inhibitory neurons, particularly those expressing Pdyn and Gal Although these mutants survive postnatally, at ∼3-5 wk they elicit a severe scratching phenotype. Behaviorally, the mutants have increased sensitivity to itch, but acute sensitivity to other sensory stimuli such as mechanical or thermal pain is unaffected. We demonstrate a requirement for positive transcriptional autoregulatory feedback to attain the level of the neuronal specification factor PTF1A necessary for generating correctly balanced neuronal circuits.


Subject(s)
Feedback, Physiological/physiology , Gene Expression Regulation/physiology , Neurons/physiology , Pruritus/genetics , Transcription Factors/genetics , Animals , CRISPR-Cas Systems , Enhancer Elements, Genetic/genetics , Mice , Mutation , Neurons/cytology , Spinal Cord , Transcription Factors/metabolism
3.
Nat Commun ; 10(1): 3201, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324758

ABSTRACT

Pulmonary neuroendocrine (NE) cancer, including small cell lung cancer (SCLC), is a particularly aggressive malignancy. The lineage-specific transcription factors Achaete-scute homolog 1 (ASCL1), NEUROD1 and POU2F3 have been reported to identify the different subtypes of pulmonary NE cancers. Using a large-scale mass spectrometric approach, here we perform quantitative secretome analysis in 13 cell lines that signify the different NE lung cancer subtypes. We quantify 1,626 proteins and identify IGFBP5 as a secreted marker for ASCL1High SCLC. ASCL1 binds to the E-box elements in IGFBP5 and directly regulates its transcription. Knockdown of ASCL1 decreases IGFBP5 expression, which, in turn, leads to hyperactivation of IGF-1R signaling. Pharmacological co-targeting of ASCL1 and IGF-1R results in markedly synergistic effects in ASCL1High SCLC in vitro and in mouse models. We expect that this secretome resource will provide the foundation for future mechanistic and biomarker discovery studies, helping to delineate the molecular underpinnings of pulmonary NE tumors.


Subject(s)
Biomarkers, Tumor , Lung Neoplasms/classification , Lung Neoplasms/metabolism , Neuroendocrine Tumors/classification , Neuroendocrine Tumors/metabolism , Transcription Factors/metabolism , Animals , Azepines/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/genetics , Octamer Transcription Factors/metabolism , Proteomics , Pyrazoles/pharmacology , Receptor, IGF Type 1/metabolism , Signal Transduction , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Triazines/pharmacology , Triazoles/pharmacology
4.
Cell Rep ; 16(5): 1259-1272, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27452466

ABSTRACT

Small cell lung carcinoma (SCLC) is a high-grade pulmonary neuroendocrine tumor. The transcription factors ASCL1 and NEUROD1 play crucial roles in promoting malignant behavior and survival of human SCLC cell lines. Here, we find that ASCL1 and NEUROD1 identify heterogeneity in SCLC, bind distinct genomic loci, and regulate mostly distinct genes. ASCL1, but not NEUROD1, is present in mouse pulmonary neuroendocrine cells, and only ASCL1 is required in vivo for tumor formation in mouse models of SCLC. ASCL1 targets oncogenic genes including MYCL1, RET, SOX2, and NFIB while NEUROD1 targets MYC. ASCL1 and NEUROD1 regulate different genes that commonly contribute to neuronal function. ASCL1 also regulates multiple genes in the NOTCH pathway including DLL3. Together, ASCL1 and NEUROD1 distinguish heterogeneity in SCLC with distinct genomic landscapes and distinct gene expression programs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Animals , Cell Line, Tumor , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Neuroendocrine Cells/metabolism , Oncogenes/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
J Thorac Oncol ; 10(4): 553-64, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25675280

ABSTRACT

INTRODUCTION: Because small-cell lung carcinomas (SCLC) are seldom resected, human materials for study are limited. Thus, genetically engineered mouse models (GEMMs) for SCLC and other high-grade lung neuroendocrine (NE) carcinomas are crucial for translational research. METHODS: The pathologies of five GEMMs were studied in detail and consensus diagnoses reached by four lung cancer pathology experts. Hematoxylin and Eosin and immunostained slides of over 100 mice were obtained from the originating and other laboratories and digitalized. The GEMMs included the original Rb/p53 double knockout (Berns Laboratory) and triple knockouts from the Sage, MacPherson, and Jacks laboratories (double knockout model plus loss of p130 [Sage laboratory] or loss of Pten [MacPherson and Jacks laboratories]). In addition, a GEMM with constitutive co-expression of SV40 large T antigen and Ascl1 under the Scgb1a1 promoter from the Linnoila laboratory were included. RESULTS: The lung tumors in all of the models had common as well as distinct pathological features. All three conditional knockout models resulted in multiple pulmonary tumors arising mainly from the central compartment (large bronchi) with foci of in situ carcinoma and NE cell hyperplasia. They consisted of inter- and intra-tumor mixtures of SCLC and large-cell NE cell carcinoma in varying proportions. Occasional adeno- or large-cell carcinomas were also seen. Extensive vascular and lymphatic invasion and metastases to the mediastinum and liver were noted, mainly of SCLC histology. In the Rb/p53/Pten triple knockout model from the MacPherson and Jacks laboratories and in the constitutive SV40/T antigen model many peripherally arising non-small-cell lung carcinoma tumors having varying degrees of NE marker expression were present (non-small-cell lung carcinoma-NE tumors). The resultant histological phenotypes were influenced by the introduction of specific genetic alterations, by inactivation of one or both alleles of specific genes, by time from Cre activation and by targeting of lung cells or NE cell subpopulations. CONCLUSION: The five GEMM models studied are representative for the entire spectrum of human high-grade NE carcinomas and are also useful for the study of multistage pathogenesis and the metastatic properties of these tumors. They represent one of the most advanced forms of currently available GEMM models for the study of human cancer.


Subject(s)
Carcinoma, Neuroendocrine/pathology , Genetic Engineering/methods , Lung Neoplasms/pathology , Lung/ultrastructure , Neoplasms, Experimental , Animals , Carcinoma, Neuroendocrine/genetics , Lung Neoplasms/genetics , Mice , Mice, Knockout , Microscopy, Electron , Tumor Cells, Cultured
6.
Mol Cell Biol ; 33(16): 3166-79, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23754747

ABSTRACT

The lineage-specific basic helix-loop-helix transcription factor Ptf1a is a critical driver for development of both the pancreas and nervous system. How one transcription factor controls diverse programs of gene expression is a fundamental question in developmental biology. To uncover molecular strategies for the program-specific functions of Ptf1a, we identified bound genomic regions in vivo during development of both tissues. Most regions bound by Ptf1a are specific to each tissue, lie near genes needed for proper formation of each tissue, and coincide with regions of open chromatin. The specificity of Ptf1a binding is encoded in the DNA surrounding the Ptf1a-bound sites, because these regions are sufficient to direct tissue-restricted reporter expression in transgenic mice. Fox and Sox factors were identified as potential lineage-specific modifiers of Ptf1a binding, since binding motifs for these factors are enriched in Ptf1a-bound regions in pancreas and neural tube, respectively. Of the Fox factors expressed during pancreatic development, Foxa2 plays a major role. Indeed, Ptf1a and Foxa2 colocalize in embryonic pancreatic chromatin and can act synergistically in cell transfection assays. Together, these findings indicate that lineage-specific chromatin landscapes likely constrain the DNA binding of Ptf1a, and they identify Fox and Sox gene families as part of this process.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Developmental , Neural Tube/embryology , Pancreas/embryology , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , Chromatin/genetics , Consensus Sequence , DNA/genetics , DNA/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Mice, Transgenic , Neural Tube/metabolism , Pancreas/metabolism , Protein Binding , SOXB1 Transcription Factors/metabolism
7.
Dev Biol ; 340(2): 283-92, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20171205

ABSTRACT

The bHLH transcription factor Neurog1 (Ngn1, Neurod3, neurogenin 1) is involved in neuronal differentiation and cell-type specification in distinct regions of the developing nervous system. Here, transgenic mouse models were developed that use a Bacterial Artificial Chromosome (BAC) containing 208kb flanking the Neurog1 gene to efficiently drive expression of GFP and Cre in all Neurog1 domains. Two characteristics of Neurog1 gene regulation were uncovered. First, a 4kb region previously shown to be sufficient for driving expression of a reporter gene to a subset of the Neurog1 pattern in the developing midbrain, hindbrain, and spinal cord is required uniformly for high levels of expression in all Neurog1 domains, even those not originally identified as being regulated by this region. Second, a 0.8 kb enhancer was identified that is sufficient to drive Neurog1-like expression specifically in the ventral neural tube. Furthermore, Neurog1 progenitor cells in the ventral neural tube are largely fated to interneuron lineages rather than to motoneurons. These studies provide new tools for directing tissue specific expression in the developing neural tube, define Neurog1 lineages in the spinal cord, and further define the complex genomic structure required for obtaining the correct levels and spatial restriction of the neuronal differentiation gene Neurog1.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Interneurons/cytology , Nerve Tissue Proteins/metabolism , Neural Tube/physiology , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage , Chromosomes, Artificial, Bacterial , Embryo, Mammalian , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Interneurons/physiology , Mice , Mice, Transgenic , Microinjections , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neural Tube/cytology , Neural Tube/embryology , Stem Cells/cytology , Transgenes/genetics
8.
Development ; 136(17): 2945-54, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19641016

ABSTRACT

PTF1-J is a trimeric transcription factor complex essential for generating the correct balance of GABAergic and glutamatergic interneurons in multiple regions of the nervous system, including the dorsal horn of the spinal cord and the cerebellum. Although the components of PTF1-J have been identified as the basic helix-loop-helix (bHLH) factor Ptf1a, its heterodimeric E-protein partner, and Rbpj, no neural targets are known for this transcription factor complex. Here we identify the neuronal differentiation gene Neurog2 (Ngn2, Math4A, neurogenin 2) as a direct target of PTF1-J. A Neurog2 dorsal neural tube enhancer localized 3' of the Neurog2 coding sequence was identified that requires a PTF1-J binding site for dorsal activity in mouse and chick neural tube. Gain and loss of Ptf1a function in vivo demonstrate its role in Neurog2 enhancer activity. Furthermore, chromatin immunoprecipitation from neural tube tissue demonstrates that Ptf1a is bound to the Neurog2 enhancer. Thus, Neurog2 expression is directly regulated by the PTF1-J complex, identifying Neurog2 as the first neural target of Ptf1a and revealing a bHLH transcription factor cascade functioning in the specification of GABAergic neurons in the dorsal spinal cord and cerebellum.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Nerve Tissue Proteins/metabolism , Spinal Cord/embryology , Transcription Factors/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Cell Differentiation/physiology , Chick Embryo , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/genetics , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/metabolism , Transcription Factors/genetics , Transcription, Genetic
9.
Dev Biol ; 328(2): 529-40, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19389376

ABSTRACT

Delta-like 3 (Dll3) is a Delta family member expressed broadly in the developing nervous system as neural progenitor cells initiate differentiation. A proximal promoter sequence for Dll3 is conserved across multiple species and is sufficient to direct GFP expression in a Dll3-like pattern in the neural tube of transgenic mice. This promoter contains multiple E-boxes, the consensus binding site for bHLH factors. Dll3 expression and the activity of the Dll3-promoter in the dorsal neural tube depends on the basic helix-loop-helix (bHLH) transcription factors Ascl1 (Mash1) and Neurog2 (Ngn2). Mutations in each E-box identified in the Dll3-promoter allowed distinct enhancer or repressor properties to be assigned to each site individually or in combination. In addition, each E-box has distinct characteristics relative to binding of bHLH factors Ascl1, Neurog1, and Neurog2. Surprisingly, novel Ascl1 containing DNA binding complexes are identified that interact with specific E-box sites within the Dll3-promoter in vitro. These complexes include Ascl1/Ascl1 homodimers and Ascl1/Neurog2 heterodimers, complexes that in some cases require additional undefined factors for efficient DNA binding. Thus, a complex interplay of E-box binding proteins spatially and temporally regulate Dll3 levels during neural tube development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Neural Tube/physiology , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Dimerization , E-Box Elements , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/genetics , Neural Tube/embryology , Neurons/physiology , Promoter Regions, Genetic , Stem Cells/physiology
10.
Development ; 134(2): 285-93, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17166924

ABSTRACT

The neural basic helix-loop-helix transcription factor Ascl1 (previously Mash1) is present in ventricular zone cells in restricted domains throughout the developing nervous system. This study uses genetic fate mapping to define the stage and neural lineages in the developing spinal cord that are derived from Ascl1-expressing cells. We find that Ascl1 is present in progenitors to both neurons and oligodendrocytes, but not astrocytes. Temporal control of the fate-mapping paradigm reveals rapid cell-cycle exit and differentiation of Ascl1-expressing cells. At embryonic day 11, Ascl1 identifies neuronal-restricted precursor cells that become dorsal horn neurons in the superficial laminae. By contrast, at embryonic day 16, Ascl1 identifies oligodendrocyte-restricted precursor cells that distribute throughout the spinal cord. These data demonstrate that sequentially generated Ascl1-expressing progenitors give rise first to dorsal horn interneurons and subsequently to late-born oligodendrocytes. Furthermore, Ascl1-null cells in the spinal cord have a diminished capacity to undergo neuronal differentiation, with a subset of these cells retaining characteristics of immature glial cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/deficiency , Cell Differentiation , DNA Primers/genetics , Female , Gene Expression Regulation, Developmental , Integrases/biosynthesis , Integrases/genetics , Interneurons/cytology , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Pregnancy , Spinal Cord/cytology , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...