Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Front Oncol ; 13: 1233953, 2023.
Article in English | MEDLINE | ID: mdl-37664059

ABSTRACT

Mutations in chromatin modifying genes frequently occur in many kinds of cancer. Most mechanistic studies focus on their canonical functions, while therapeutic approaches target their enzymatic activity. Recent studies, however, demonstrate that non-canonical functions of chromatin modifiers may be equally important and therapeutically actionable in different types of cancer. One epigenetic regulator that demonstrates such a dual role in cancer is the histone methyltransferase EZH2. EZH2 is a core component of the polycomb repressive complex 2 (PRC2), which plays a crucial role in cell identity, differentiation, proliferation, stemness and plasticity. While much of the regulatory functions and oncogenic activity of EZH2 have been attributed to its canonical, enzymatic activity of methylating lysine 27 on histone 3 (H3K27me3), a repressive chromatin mark, recent studies suggest that non-canonical functions that are independent of H3K27me3 also contribute towards the oncogenic activity of EZH2. Contrary to PRC2's canonical repressive activity, mediated by H3K27me3, outside of the complex EZH2 can directly interact with transcription factors and oncogenes to activate gene expression. A more focused investigation into these non-canonical interactions of EZH2 and other epigenetic/chromatin regulators may uncover new and more effective therapeutic strategies. Here, we summarize major findings on the non-canonical functions of EZH2 and how they are related to different aspects of carcinogenesis.

3.
Oncogene ; 41(46): 4983-4993, 2022 11.
Article in English | MEDLINE | ID: mdl-36220978

ABSTRACT

Enhancer of Zeste Homolog 2 (EZH2) is the catalytic component of the Polycomb Repressive Complex 2, a chromatin modifying complex, which mediates methylation of lysine 27 on histone 3 (H3K27me3), a repressive chromatin mark. Genetic alterations in EZH2 in melanoma include amplifications and activating point mutations at tyrosine 641 (Y641) whose underlying oncogenic mechanisms remain largely unknown. Here, we found that expression of Ezh2Y641F causes upregulation of a subset of interferon-regulated genes in melanoma cells. Upregulation of these genes was not a direct effect of changes in H3K27me3, but via a non-canonical interaction between Ezh2 and Signal Transducer and Activator of Transcription 3 (Stat3). Ezh2 and Stat3 together function as transcriptional activators to mediate gene activation of numerous genes, including MHC Class 1b antigen processing genes. Furthermore, expression of Stat3 is required to maintain an anti-tumor immune response in Ezh2Y641F melanomas and to prevent melanoma progression and recurrence.


Subject(s)
Enhancer of Zeste Homolog 2 Protein , Melanoma , Humans , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Histones/genetics , Histones/metabolism , Antigen Presentation , Mutation , Melanoma/genetics , Chromatin/genetics
4.
Clin Lymphoma Myeloma Leuk ; 19(11): 744-750, 2019 11.
Article in English | MEDLINE | ID: mdl-31551170

ABSTRACT

BACKGROUND: EZH2 is a histone methyltransferase that suppresses genes involved in cell cycle control. Overexpression of EZH2 has been associated with a poor prognosis in various malignancies. Pawlyn et al recently reported poor outcomes in patients with multiple myeloma and overexpression of EZH2. In order to validate these findings, we analyzed EZH2 expression and outcomes among patients from the CoMMpass study. PATIENTS AND METHODS: We extracted clinical, expression, and genomic data from Interim Analysis 13 of the MMRF CoMMpass study, which harbors data from over 1000 patients with multiple myeloma. Correlations were drawn between EZH2 expression and common genetic mutations. We analyzed the association of EZH2 overexpression with progression-free (PFS) and overall survival (OS). RESULTS: The estimated median PFS for patients with EZH2 overexpression was 20.2 months (95% confidence interval [CI], 16.3-25.5 months) compared with 37.2 months (95% CI, 31.5-40.7 months) for patients without (P < .001). The estimated median OS for patients with EZH2 overexpression was 52.3 months (95% CI, 38.5 months to unable to quantitate), whereas the median OS had not been reached for those without (P < .001). EZH2 overexpression was more common in those with 17p and 1q deletions, TP53 missense mutations, and certain KRAS mutations. Coinciding BRAF and EZH2 amplification occurred frequently. CONCLUSION: EZH2 overexpression is associated with worse outcomes among patients with multiple myeloma from the CoMMpass study. Its known association with p53 and other drivers of malignancy support further lab-based and clinical study in multiple myeloma.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Adult , Aged , Aged, 80 and over , Alleles , Biomarkers, Tumor , Female , Humans , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Mutation , Neoplasm Staging , Prognosis , Survival Analysis
5.
Proc Natl Acad Sci U S A ; 116(7): 2603-2611, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30683717

ABSTRACT

The activation of cellular senescence throughout the lifespan promotes tumor suppression, whereas the persistence of senescent cells contributes to aspects of aging. This theory has been limited, however, by an inability to identify and isolate individual senescent cells within an intact organism. Toward that end, we generated a murine reporter strain by "knocking-in" a fluorochrome, tandem-dimer Tomato (tdTom), into exon 1α of the p16INK4a locus. We used this allele (p16tdTom ) for the enumeration, isolation, and characterization of individual p16INK4a -expressing cells (tdTom+). The half-life of the knocked-in transcript was shorter than that of the endogenous p16INK4a mRNA, and therefore reporter expression better correlated with p16INK4a promoter activation than p16INK4a transcript abundance. The frequency of tdTom+ cells increased with serial passage in cultured murine embryo fibroblasts from p16tdTom/+ mice. In adult mice, tdTom+ cells could be readily detected at low frequency in many tissues, and the frequency of these cells increased with aging. Using an in vivo model of peritoneal inflammation, we compared the phenotype of cells with or without activation of p16INK4a and found that tdTom+ macrophages exhibited some features of senescence, including reduced proliferation, senescence-associated ß-galactosidase (SA-ß-gal) activation, and increased mRNA expression of a subset of transcripts encoding factors involved in SA-secretory phenotype (SASP). These results indicate that cells harboring activation of the p16INK4a promoter accumulate with aging and inflammation in vivo, and display characteristics of senescence.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Promoter Regions, Genetic , Animals , Cell Proliferation , Enzyme Activation , Fibroblasts/metabolism , Half-Life , Humans , Mice , Phenotype , RNA, Messenger/genetics , beta-Galactosidase/metabolism
6.
Exp Hematol ; 51: 63-70.e1, 2017 07.
Article in English | MEDLINE | ID: mdl-28435024

ABSTRACT

LKB1 (also known as STK11) is a potent tumor suppressor in solid tumors, such as melanoma and lung adenocarcinoma, but inactivation in hematopoietic cells causes cell death without signs of tumorigenesis. We noted somatic LKB1 deletion or mutation at low frequency in human B-cell lymphoma. To determine if LKB1 inactivation is a passenger or driver event in lymphoid cancers, we examined the effects of conditional inactivation of Lkb1 in murine lymphocytes. Consistent with prior reports, Lkb1 deletion in either T or B cells resulted in massive, lineage-specific apoptosis. Surprisingly, despite an 80% reduction of peripheral B-cell number, animals harboring somatic B-lineage Lkb1 deletion developed aggressive B-cell lymphoma with high penetrance and moderate latency. Malignant B cells exhibited somatic Lkb1 recombination. In contrast, Lkb1 deletion in T cells did not promote tumorigenesis. Concomitant Ras activation with Lkb1 deletion reduced T-cell apoptosis, but did not enhance tumor formation in T or B cells. These results suggest that although physiologic LKB1 expression exerts a potent pro-survival effect in lymphocytes, LKB1 inactivation nonetheless facilitates transformation of B, but not T, lymphocytes.


Subject(s)
Apoptosis/genetics , B-Lymphocytes/metabolism , Cell Transformation, Neoplastic , Gene Deletion , Lymphoma, B-Cell , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinases , Animals , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Mice , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
7.
Nat Med ; 22(6): 632-40, 2016 06.
Article in English | MEDLINE | ID: mdl-27135738

ABSTRACT

B cell lymphoma and melanoma harbor recurrent mutations in the gene encoding the EZH2 histone methyltransferase (EZH2), but the carcinogenic role of these mutations is unclear. Here we describe a mouse model in which the most common somatic Ezh2 gain-of-function mutation (EZH2(Y646F) in human; Ezh2(Y641F) in mouse) is conditionally expressed. Expression of Ezh2(Y641F) in mouse B cells or melanocytes caused high-penetrance lymphoma or melanoma, respectively. Overexpression of the anti-apoptotic protein Bcl2, but not the oncoprotein Myc, or loss of the tumor suppressor protein p53 (encoded by Trp53 in mice) further accelerated lymphoma progression. Expression of the mutant Braf but not the mutant Nras oncoprotein further accelerated melanoma progression. Although expression of Ezh2(Y641F) globally increased the abundance of trimethylated Lys27 of histone H3 (H3K27me3), it also caused a widespread redistribution of this repressive mark, including a loss of H3K27me3 that was associated with increased transcription at many loci. These results suggest that Ezh2(Y641F) induces lymphoma and melanoma through a vast reorganization of chromatin structure, inducing both repression and activation of polycomb-regulated loci.


Subject(s)
Carcinogenesis/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic/genetics , Histone Code/genetics , Lymphoma, B-Cell/genetics , Melanoma/genetics , Animals , B-Lymphocytes/metabolism , Cell Line, Tumor , Disease Progression , Gene Knockdown Techniques , Humans , Immunoblotting , Melanocytes/metabolism , Methylation , Mice , Monomeric GTP-Binding Proteins/genetics , Mutation , Neoplasm Transplantation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/genetics , Real-Time Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics
8.
Comp Med ; 65(3): 165-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26141441

ABSTRACT

Mice are used extensively in transplantation studies involving bone marrow ablation. Due to the increasing security issues and expenses involved with γ irradiators, self-contained X-ray irradiators have been increasing in popularity. We hypothesized that bone marrow ablation by irradiation of mice with a (137)Cs irradiator would be comparable to that from an X-ray source irradiator. A lethal-dose curve was obtained by irradiating C57BL/6J mice with 500, 700, 900, and 1100 cGy from either source. These data were used to determine the lethal radiation exposure range for a noncompetitive bone marrow engraftment curve for each source. At 90 d after reconstitution, the bone marrow engraftment curves revealed significant differences between the 2 sources in the establishment of B cell, myeloid, and T cell lineages. Murine B cell reconstitution after exposure to a (137)Cs source was greater than that after X-ray exposure at each dose level, whereas the converse was true for myeloid cell reconstitution. At the 1050- and 1100-cGy doses, mice irradiated by using the X-ray source demonstrated higher levels of T cell reconstitution but decreased survival compared with mice irradiated with the (137)Cs source. We concluded that although both sources ablated endogenous bone marrow sufficiently to enable stem cell engraftment, there are distinct physiologic responses that should be considered when choosing the optimal source for use in a study and that irradiation from the (137)Cs source was associated with lower overall morbidity due to opportunistic infection.


Subject(s)
Bone Marrow Transplantation , Cesium Radioisotopes/administration & dosage , Transplantation Conditioning , X-Rays , Animals , Lethal Dose 50 , Male , Mice , Mice, Inbred C57BL
9.
Cancer Cell ; 27(1): 3-5, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25584887

ABSTRACT

Oncogene-induced senescence (OIS) is thought to be a barrier to malignant transformation resulting from the strong activation of oncogenes. In this issue of Cancer Cell, Damsky and colleagues suggest activation of mTORC1 and mTORC2 is required for OIS evasion in human melanomas harboring oncogenic BRAF mutations.


Subject(s)
Melanoma, Experimental/pathology , Multiprotein Complexes/metabolism , Nevus/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Humans , Mechanistic Target of Rapamycin Complex 1
10.
Cancer Discov ; 4(12): 1418-29, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25252692

ABSTRACT

UNLABELLED: NRAS mutation at codons 12, 13, or 61 is associated with transformation; yet, in melanoma, such alterations are nearly exclusive to codon 61. Here, we compared the melanoma susceptibility of an NrasQ61R knock-in allele to similarly designed KrasG12D and NrasG12D alleles. With concomitant p16INK4a inactivation, KrasG12D or NrasQ61R expression efficiently promoted melanoma in vivo, whereas NrasG12D did not. In addition, NrasQ61R mutation potently cooperated with Lkb1/Stk11 loss to induce highly metastatic disease. Functional comparisons of NrasQ61R and NrasG12D revealed little difference in the ability of these proteins to engage PI3K or RAF. Instead, NrasQ61R showed enhanced nucleotide binding, decreased intrinsic GTPase activity, and increased stability when compared with NrasG12D. This work identifies a faithful model of human NRAS-mutant melanoma, and suggests that the increased melanomagenecity of NrasQ61R over NrasG12D is due to heightened abundance of the active, GTP-bound form rather than differences in the engagement of downstream effector pathways. SIGNIFICANCE: This work explains the curious predominance in human melanoma of mutations of codon 61 of NRAS over other oncogenic NRAS mutations. Using conditional "knock-in" mouse models, we show that physiologic expression of NRASQ61R, but not NRASG12D, drives melanoma formation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Codon , Genes, ras , Melanoma/genetics , Mutation , AMP-Activated Protein Kinase Kinases , Alleles , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Deletion , Gene Order , Genetic Loci , Genotype , Guanosine Triphosphate/metabolism , Humans , Melanoma/metabolism , Melanoma/mortality , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Metastasis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Tumor Burden
12.
PLoS Genet ; 9(3): e1003359, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555280

ABSTRACT

Hematopoietic stem cells (HSCs) are rare quiescent cells that continuously replenish the cellular components of the peripheral blood. Observing that the ataxia-associated gene Ataxin-1-like (Atxn1L) was highly expressed in HSCs, we examined its role in HSC function through in vitro and in vivo assays. Mice lacking Atxn1L had greater numbers of HSCs that regenerated the blood more quickly than their wild-type counterparts. Molecular analyses indicated Atxn1L null HSCs had gene expression changes that regulate a program consistent with their higher level of proliferation, suggesting that Atxn1L is a novel regulator of HSC quiescence. To determine if additional brain-associated genes were candidates for hematologic regulation, we examined genes encoding proteins from autism- and ataxia-associated protein-protein interaction networks for their representation in hematopoietic cell populations. The interactomes were found to be highly enriched for proteins encoded by genes specifically expressed in HSCs relative to their differentiated progeny. Our data suggest a heretofore unappreciated similarity between regulatory modules in the brain and HSCs, offering a new strategy for novel gene discovery in both systems.


Subject(s)
Ataxia , Autistic Disorder , Gene Regulatory Networks , Hematopoietic Stem Cells , Nerve Tissue Proteins , Nuclear Proteins , Animals , Ataxia/genetics , Ataxia/metabolism , Ataxin-1 , Ataxins , Autistic Disorder/genetics , Autistic Disorder/metabolism , Brain/metabolism , Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Genetic Association Studies , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity , Protein Interaction Maps
13.
Nat Immunol ; 13(8): 761-9, 2012 Jul 08.
Article in English | MEDLINE | ID: mdl-22772404

ABSTRACT

Thymopoiesis depends on the recruitment and expansion of bone marrow-derived progenitor populations; tight regulation of these processes is required for maintenance of the homeostasis of the T lineage. Lyl-1, a transcription factor that regulates hematopoietic progenitors, is expressed in thymocyte progenitors until T cell commitment. Here we demonstrate a requirement for Lyl-1 in lymphoid specification and the maintenance of early T lineage progenitors (ETPs). Lyl-1 deficiency resulted in profound defects in the generation of lymphoid-primed multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs) and ETPs. Lyl-1-deficient ETPs and thymocyte progenitors at the CD4(-)CD8(-) double-negative 2 (DN2) stage showed more apoptosis, blocked differentiation and impaired population expansion. We identified Gfi1 as a critical transcriptional target of Lyl-1-mediated lymphopoiesis of T cells. Thus, Lyl-1 is a pivotal component of a transcriptional program that controls the lymphoid specification and maintenance of ETPs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Lymphoid Progenitor Cells/physiology , Lymphopoiesis , Neoplasm Proteins/metabolism , T-Lymphocytes/immunology , Animals , Apoptosis/immunology , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Marrow Cells/physiology , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Lineage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Lymphoid Progenitor Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , T-Lymphocytes/physiology , Thymocytes/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Genesis ; 49(6): 441-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21387538

ABSTRACT

Lyl1 codes for a bHLH protein that is an important regulator of hematopoietic stem cell function. An existing mutant allele of Lyl1 features a lacZ gene inserted in-frame into the fourth exon, leaving behind the N-terminus and the DNA-binding basic region, resulting in a translated chimeric protein. Here, we have generated a null allele, which allowed us to examine residual function of the N-terminus in the absence of a bHLH region. The new Lyl1-/- mouse model exhibited a reduced ability to generate lymphoid lineages and a somewhat more severe hematopoietic repopulation defect when transplanting purified hematopoietic stem cells. Our data show that in the absence of the HLH but presence of the N-terminus, residual function of the Lyl1 is detectable but relatively minor. The new model may be of use for studies of Lyl1 in which a null allele is required, or for which presence of the LacZ may complicate the combined use of additional mouse models bearing the lacZ marker.


Subject(s)
Alleles , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Hematopoietic Stem Cells/physiology , Models, Animal , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Animals , Blood Cell Count , Blotting, Southern , Bone Marrow Transplantation , DNA Primers/genetics , Flow Cytometry , Genotype , Mice , Mice, Knockout/genetics
15.
Leuk Res ; 35(3): 405-12, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20705338

ABSTRACT

This study investigates the function of the lymphoblastic leukemia gene, Lyl1 in the hematopoietic system and its oncogenic potential in the development of leukemia. Overexpression of Lyl1 in mouse bone marrow cells caused T-cell increase in the peripheral blood and expansion of the hematopoietic progenitors in culture and in the bone marrow. These observations were the result of increased proliferation and suppressed apoptosis of the progenitor cells caused by the Lyl1-overexpression. Our studies present substantial evidence supporting the secondary, pro-leukemic effect of Lyl1 in early hematopoietic progenitors with the potential to cause expansion of malignant cells with a stem/early progenitor-like phenotype.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Leukemia/pathology , Neoplasm Proteins/physiology , T-Lymphocytes/pathology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Apoptosis , Blotting, Western , Bone Marrow Transplantation , Cell Proliferation , Colony-Forming Units Assay , Hematopoietic System , Leukemia/immunology , Leukemia/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , RNA, Messenger/genetics , Receptor, Notch1 , Reverse Transcriptase Polymerase Chain Reaction
16.
Cell Stem Cell ; 4(2): 180-6, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19200805

ABSTRACT

Scl and Lyl1 encode two related basic-helix-loop-helix transcription factors implicated in T cell acute lymphoblastic leukemia. Previous studies showed that Scl is essential for embryonic and adult erythropoiesis, while Lyl1 is important for B cell development. Single-knockout mice have not revealed an essential function for Scl or Lyl1 in adult hematopoietic stem cells (HSCs). To determine if maintenance of HSCs in single-knockout mice is due to functional redundancy, we generated Lyl1;Scl-conditional double-knockout mice. Here, we report a striking genetic interaction between the two genes, with a clear dose dependence for the presence of Scl or Lyl1 alleles for HSC function. Bone marrow repopulation assays and analyses demonstrated rapid loss of hematopoietic progenitors due to apoptosis. The function of HSCs could be rescued by a single allele of Lyl1 but not Scl. These results show that expression of at least one of these factors is essential for maintenance of adult HSC function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival , Hematopoietic Stem Cells/physiology , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Stem Cells/physiology , Alleles , Animals , Apoptosis/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Marrow Transplantation , Cells, Cultured , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Radiation, Ionizing , Stem Cells/cytology , T-Cell Acute Lymphocytic Leukemia Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...