Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Nat Commun ; 15(1): 566, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38263303

ABSTRACT

Complexity of quantum phases of matter is often understood theoretically by using gauge structures, as is recognized by the [Formula: see text] and U(1) gauge theory description of spin liquids in frustrated magnets. Anomalous Hall effect of conducting electrons can intrinsically arise from a U(1) gauge expressing the spatial modulation of ferromagnetic moments or from an SU(2) gauge representing the spin-orbit coupling effect. Similarly, in insulating ferro and antiferromagnets, the magnon contribution to anomalous transports is explained in terms of U(1) and SU(2) fluxes present in the ordered magnetic structure. Here, we report thermal Hall measurements of MnSc2S4 in an applied field up to 14 T, for which we consider an emergent higher rank SU(3) flux, controlling the magnon transport. The thermal Hall coefficient takes a substantial value when the material enters a three-sublattice antiferromagnetic skyrmion phase, which is in agreement with the linear spin-wave theory. In our description, magnons are dressed with SU(3) gauge field, which is a mixture of three species of U(1) gauge fields originating from the slowly varying magnetic moments on these sublattices.

2.
Rev Sci Instrum ; 94(11)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37938066

ABSTRACT

We developed a metallic pressure cell made of 56Ni-40Cr-4Al (Ni-Cr-Al) alloy for use with a non-destructive pulse magnet and a magnetic susceptibility measurement apparatus with a proximity detector oscillator (PDO) in pulsed magnetic fields of up to 51 T under pressures of up to 2.1 GPa. Both the sample and sensor coil of the PDO were placed in the cell so that the magnetic signal from Ni-Cr-Al would not overlay the intrinsic magnetic susceptibility of the sample. A systematic investigation of the Joule heating originating from metallic parts of the pressure cell revealed that the increase in sample temperature is negligible at 1.4 K in magnetic fields of up to 40 T in the field-ascending process for the maximum applied magnetic field of 51 T. The effectiveness of our apparatus was demonstrated by investigating the pressure dependence of the magnetization process of the triangular-lattice antiferromagnet Ba3CoSb2O9.

3.
Sci Rep ; 12(1): 17287, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36241850

ABSTRACT

We developed a new type of compact magnetic force booster by which we succeeded in crystallizing proteins (hen egg white lysozyme) while making them levitate in a solution without contacting the container. This technique is noteworthy in the practical merit that we could control the growth of crystals from the initial stage of nucleation in a magnetic field of merely a few Tesla. The shape of the booster was designed in accordance with the dynamical stability against external forces acting on the crystals. Under a stable condition, the crystals condensed spherically, and formed a "shell shaped" crystallization with a hollow interior. Our magnetic force booster has the potential for use in innovation, especially in the field of protein crystal engineering.


Subject(s)
Chickens , Muramidase , Animals , Crystallization/methods , Magnetic Phenomena , Muramidase/chemistry , Proteins
4.
Cancers (Basel) ; 14(11)2022 May 24.
Article in English | MEDLINE | ID: mdl-35681561

ABSTRACT

The MUC1-C apical transmembrane protein is activated in the acute response of epithelial cells to inflammation. However, chronic MUC1-C activation promotes cancer progression, emphasizing the importance of MUC1-C as a target for treatment. We report here that MUC1-C is necessary for intrinsic expression of the RIG-I, MDA5 and cGAS cytosolic nucleotide pattern recognition receptors (PRRs) and the cGAS-stimulator of IFN genes (STING) in triple-negative breast cancer (TNBC) cells. Consistent with inducing the PRR/STING axis, MUC1-C drives chronic IFN-ß production and activation of the type I interferon (IFN) pathway. MUC1-C thereby induces the IFN-related DNA damage resistance gene signature (IRDS), which includes ISG15, in linking chronic inflammation with DNA damage resistance. Targeting MUC1-C in TNBC cells treated with carboplatin or the PARP inhibitor olaparib further demonstrated that MUC1-C is necessary for expression of PRRs, STING and ISG15 and for intrinsic DNA damage resistance. Of translational relevance, MUC1 significantly associates with upregulation of STING and ISG15 in TNBC tumors and is a target for treatment with CAR T cells, antibody-drug conjugates (ADCs) and direct inhibitors that are under preclinical and clinical development.

5.
Oncogene ; 41(27): 3511-3523, 2022 07.
Article in English | MEDLINE | ID: mdl-35688945

ABSTRACT

Merkel cell carcinoma (MCC) is an aggressive malignancy with neuroendocrine (NE) features, limited treatment options, and a lack of druggable targets. There is no reported involvement of the MUC1-C oncogenic protein in MCC progression. We show here that MUC1-C is broadly expressed in MCCs and at higher levels in Merkel cell polyomavirus (MCPyV)-positive (MCCP) relative to MCPyV-negative (MCCN) tumors. Our results further demonstrate that MUC1-C is expressed in MCCP, as well as MCCN, cell lines and regulates common sets of signaling pathways related to RNA synthesis, processing, and transport in both subtypes. Mechanistically, MUC1-C (i) interacts with MYCL, which drives MCC progression, (ii) is necessary for expression of the OCT4, SOX2, KLF4, MYC, and NANOG pluripotency factors, and (iii) induces the NEUROD1, BRN2 and ATOH1 NE lineage dictating transcription factors. We show that MUC1-C is also necessary for MCCP and MCCN cell survival by suppressing DNA replication stress, the p53 pathway, and apoptosis. In concert with these results, targeting MUC1-C genetically and pharmacologically inhibits MCC self-renewal capacity and tumorigenicity. These findings demonstrate that MCCP and MCCN cells are addicted to MUC1-C and identify MUC1-C as a potential target for MCC treatment.


Subject(s)
Carcinoma, Merkel Cell , Merkel cell polyomavirus , Mucin-1 , Skin Neoplasms , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/virology , Humans , Mucin-1/metabolism , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/virology
6.
Mol Cancer Res ; 20(9): 1379-1390, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35612556

ABSTRACT

Small cell lung cancer (SCLC) is a recalcitrant malignancy defined by subtypes on the basis of differential expression of the ASCL1, NEUROD1, and POU2F3 transcription factors. The MUC1-C protein is activated in pulmonary epithelial cells by exposure to environmental carcinogens and promotes oncogenesis; however, there is no known association between MUC1-C and SCLC. We report that MUC1-C is expressed in classic neuroendocrine (NE) SCLC-A, variant NE SCLC-N and non-NE SCLC-P cells and activates the MYC pathway in these subtypes. In SCLC cells characterized by NE differentiation and DNA replication stress, we show that MUC1-C activates the MYC pathway in association with induction of E2F target genes and dysregulation of mitotic progression. Our studies further demonstrate that the MUC1-C→MYC pathway is necessary for induction of (i) NOTCH2, a marker of pulmonary NE stem cells that are the proposed cell of SCLC origin, and (ii) ASCL1 and NEUROD1. We also show that the MUC1-C→MYC→NOTCH2 network is necessary for self-renewal capacity and tumorigenicity of NE and non-NE SCLC cells. Analyses of datasets from SCLC tumors confirmed that MUC1 expression in single SCLC cells significantly associates with activation of the MYC pathway. These findings demonstrate that SCLC cells are addicted to MUC1-C and identify a potential new target for SCLC treatment. IMPLICATIONS: This work uncovers addiction of SCLC cells to MUC1-C, which is a druggable target that could provide new opportunities for advancing SCLC treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Neuroendocrine Cells , Small Cell Lung Carcinoma , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Mucin-1/genetics , Mucin-1/metabolism , Neuroendocrine Cells/pathology , Oncogene Proteins/genetics , Small Cell Lung Carcinoma/genetics
7.
Oncoimmunology ; 11(1): 2029298, 2022.
Article in English | MEDLINE | ID: mdl-35127252

ABSTRACT

The oncogenic MUC1-C protein drives dedifferentiation of castrate resistant prostate cancer (CRPC) cells in association with chromatin remodeling. The present work demonstrates that MUC1-C is necessary for expression of IFNGR1 and activation of the type II interferon-gamma (IFN-γ) pathway. We show that MUC1-C→ARID1A/BAF signaling induces IFNGR1 transcription and that MUC1-C-induced activation of the NuRD complex suppresses FBXW7 in stabilizing the IFNGR1 protein. MUC1-C and NuRD were also necessary for expression of the downstream STAT1 and IRF1 transcription factors. We further demonstrate that MUC1-C and PBRM1/PBAF are necessary for IRF1-induced expression of (i) IDO1, WARS and PTGES, which metabolically suppress the immune tumor microenvironment (TME), and (ii) the ISG15 and SERPINB9 inhibitors of T cell function. Of translational relevance, we show that MUC1 associates with expression of IFNGR1, STAT1 and IRF1, as well as the downstream IDO1, WARS, PTGES, ISG15 and SERPINB9 immunosuppressive effectors in CRPC tumors. Analyses of scRNA-seq data further demonstrate that MUC1 correlates with cancer stem cell (CSC) and IFN gene signatures across CRPC cells. Consistent with these results, MUC1 associates with immune cell-depleted "cold" CRPC TMEs. These findings demonstrate that MUC1-C integrates chronic activation of the type II IFN-γ pathway and induction of chromatin remodeling complexes in linking the CSC state with immune evasion.


Subject(s)
Chromatin Assembly and Disassembly , Interferon-gamma , Mucin-1 , Prostatic Neoplasms, Castration-Resistant , Chromatin Assembly and Disassembly/immunology , Humans , Immunosuppression Therapy , Male , Mucin-1/immunology , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Interferon/immunology , Signal Transduction/immunology , Tumor Microenvironment , Interferon gamma Receptor
8.
Mol Cancer Res ; 20(4): 556-567, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35022313

ABSTRACT

The oncogenic MUC1-C protein promotes dedifferentiation of castrate-resistant prostate cancer (CRPC) and triple-negative breast cancer (TNBC) cells. Chromatin remodeling is critical for the cancer stem cell (CSC) state; however, there is no definitive evidence that MUC1-C regulates chromatin accessibility and thereby expression of stemness-associated genes. We demonstrate that MUC1-C drives global changes in chromatin architecture in the dedifferentiation of CRPC and TNBC cells. Our results show that MUC1-C induces differentially accessible regions (DAR) across their genomes, which are significantly associated with differentially expressed genes (DEG). Motif and cistrome analysis further demonstrated MUC1-C-induced DARs align with genes regulated by the JUN/AP-1 family of transcription factors. MUC1-C activates the BAF chromatin remodeling complex, which is recruited by JUN in enhancer selection. In studies of the NOTCH1 gene, which is required for CRPC and TNBC cell self-renewal, we demonstrate that MUC1-C is necessary for (i) occupancy of JUN and ARID1A/BAF, (ii) increases in H3K27ac and H3K4me3 signals, and (iii) opening of chromatin accessibility on a proximal enhancer-like signature. Studies of the EGR1 and LY6E stemness-associated genes further demonstrate that MUC1-C-induced JUN/ARID1A complexes regulate chromatin accessibility on proximal and distal enhancer-like signatures. These findings uncover a role for MUC1-C in chromatin remodeling that is mediated at least in part by JUN/AP-1 and ARID1A/BAF in association with driving the CSC state. IMPLICATIONS: These findings show that MUC1-C, which is necessary for the CRPC and TNBC CSC state, activates a novel pathway involving JUN/AP-1 and ARID1A/BAF that regulates chromatin accessibility of stemness-associated gene enhancers.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation, Neoplastic , Carcinogenesis/genetics , Chromatin/genetics , Chromatin/metabolism , Humans , Male , Mucin-1/metabolism , Neoplastic Stem Cells/metabolism , Oncogenes
9.
Eur Urol ; 81(2): 145-148, 2022 02.
Article in English | MEDLINE | ID: mdl-34627641

ABSTRACT

Immune checkpoint inhibitors (ICIs) have become key agents in the management of clear cell renal cell carcinoma (ccRCC), but their benefits are limited and responders remain unidentified. We investigated the significance of PARP1 in ccRCC using RNA sequencing data for 311 tumors from patients enrolled in prospective clinical trials of PD-1 blockade. Among patients treated with nivolumab (n = 181), overall survival (OS) was significantly higher in the PARP1-low group than in the PARP1-high group (p = 0.006), and PARP1 status was significantly associated with OS (hazard ratio [HR] 1.7; p = 0.007). By contrast, for patients treated with everolimus (n = 130) there was no significant difference by PARP1 status for progression-free survival (PFS; p = 0.9) or OS (p = 0.38). In subgroup analysis for PBRM1-mutated ccRCC, PFS (p = 0.016) and OS (p = 0.004) were significantly longer in the group with PARP1-low status and PBRM1 mutation in comparison to the other groups. In addition, PARP1 status was significantly associated with PFS (HR 2.6; p = 0.007) and OS (HR 3.5; p = 0.016) among patients with PBRM1-mutated ccRCC treated with nivolumab. Our study suggests that PARP1 can be used as a biomarker for predicting response to ICI treatment for patients with PBRM1-mutated ccRCC. PATIENT SUMMARY: Immune checkpoint inhibitors (ICIs) are key agents in the treatment of multiple cancers. We found that expression of the PARP1 protein was associated with survival after ICI treatment and with the response to ICI treatment in patients with clear cell kidney cancer who have a mutation of the PBRM1 gene.


Subject(s)
Carcinoma, Renal Cell , Carcinoma , Kidney Neoplasms , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biomarkers , Carcinoma/drug therapy , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , DNA-Binding Proteins/genetics , Female , Humans , Immune Checkpoint Inhibitors , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Male , Nivolumab/therapeutic use , Poly (ADP-Ribose) Polymerase-1/genetics , Prospective Studies , Transcription Factors
10.
Carcinogenesis ; 43(1): 67-76, 2022 02 11.
Article in English | MEDLINE | ID: mdl-34657147

ABSTRACT

Pancreatic ductal adenocarcinomas (PDAC) and poorly differentiated pancreatic neuroendocrine (NE) carcinomas are KRAS mutant malignancies with a potential common cell of origin. PDAC ductal, but not NE, lineage traits have been associated with cell-intrinsic activation of interferon (IFN) pathways. The present studies demonstrate that the MUC1 C-terminal subunit (MUC1-C), which evolved to protect mammalian epithelia from loss of homeostasis, is aberrantly overexpressed in KRAS mutant PDAC tumors and cell lines. We show that MUC1-C is necessary for activation of the type I and II IFN pathways and for expression of the Yamanaka OCT4, SOX2, KLF4 and MYC (OSKM) pluripotency factors. Our results demonstrate that MUC1-C integrates IFN signaling and pluripotency with NE dedifferentiation by forming a complex with MYC and driving the (i) achaete-scute homolog 1 and BRN2/POU3F2 neural, and (ii) NOTCH1/2 stemness transcription factors. Of translational relevance, targeting MUC1-C genetically and pharmacologically in PDAC cells (i) suppresses OSKM, NE dedifferentiation and NOTCH1/2, and (ii) inhibits self-renewal capacity and tumorigenicity. In PDAC tumors, we show that MUC1 significantly associates with activation of IFN signaling, MYC and NOTCH, and that upregulation of the MUC1-C → MYC pathway confers a poor prognosis. These findings indicate that MUC1-C dictates PDAC NE lineage specification and is a potential target for the treatment of recalcitrant pancreatic carcinomas with NE dedifferentiation.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Mucin-1/genetics , Neuroendocrine Cells/pathology , Pancreatic Neoplasms/genetics , Adenocarcinoma/pathology , Animals , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice, Nude , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Signal Transduction/genetics , Pancreatic Neoplasms
11.
Oncogene ; 40(30): 4930-4940, 2021 07.
Article in English | MEDLINE | ID: mdl-34163028

ABSTRACT

The polybromo-associated PBAF (SWI/SNF) chromatin remodeling complex, which includes PBRM1, ARID2, and BRD7, regulates cell differentiation and genomic integrity. MUC1-C is an oncogenic protein that drives lineage plasticity in prostate cancer (PC) progression. The present work demonstrates that MUC1-C induces PBRM1, ARID2, and BRD7 expression by the previously unrecognized E2F1-mediated activation of their respective promoters. The functional significance of the MUC1-C→PBAF pathway is supported by demonstrating involvement of MUC1-C in associating with nuclear PBAF and driving the NRF2 antioxidant gene transcriptome in PC cells. Mechanistically, MUC1-C forms a complex with NRF2 and PBRM1 on the NRF2 target SLC7A11 gene that encodes the xCT cystine-glutamate antiporter, increases chromatin accessibility and induces SLC7A11/xCT expression. We also show that MUC1-C and PBRM1 are necessary for induction of other NRF2 target genes, including G6PD and PGD that regulate the pentose phosphate pathway. Our results further demonstrate that MUC1-C integrates activation of PBRM1 with the regulation of antioxidant genes, ROS levels, pluripotency factor expression and the cancer stem cell (CSC) state. These findings reveal a role for MUC1-C in regulating PBAF, redox balance and lineage plasticity of PC CSC progression. Our findings also uncover involvement of MUC1-C in integrating the PBAF and BAF pathways in cancer.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Mucin-1/metabolism , Neoplastic Stem Cells/metabolism , Oxidation-Reduction , Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Biomarkers , Biomarkers, Tumor , Cell Line , Disease Susceptibility , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Humans , Male , Models, Biological , Oxidative Stress , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , Protein Binding , Signal Transduction
12.
Dalton Trans ; 50(15): 5251-5261, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33881078

ABSTRACT

Two mononuclear complexes [Ni(dapsc)(H2O)2]Cl(NO3)·H2O (1) and [Ni(dapsc)(NCS)2] (2), and a bimetallic CN-bridged trinuclear molecule [NiII(dapsc)(H2O)]2[WIV(CN)8]·11H2O (3) (dapsc = 2,6-diacetylpyridine-bis(semicarbazone)) were synthesised and characterised in terms of structure and magnetic properties. All three compounds contain Ni(ii) ions in a pentagonal bipyramid coordination geometry afforded by the equatorial pentadentate ligand (dapsc) and two O- or N-donating axial ligands. The compounds differ in the relative arrangement of the complexes, intermolecular interactions and distortion from the ideal coordination geometry. The high-field EPR and magnetometric studies show large anisotropy of the Ni(ii) centres with the D parameters in the range of -10.5 to -21.2 cm-1 and negligible antiferromagnetic interactions. The easy-axis magnetic anisotropies of 1-3 were reproduced by ab initio CASSCF/NEVPT2 calculations. The ground states consist mainly of the |MS = |±1 states, which is consistent with the fact that no out-of-phase signal can be detected in the AC magnetic susceptibility measurements.

13.
J Immunother Cancer ; 9(1)2021 01.
Article in English | MEDLINE | ID: mdl-33495298

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) have had a profound impact on the treatment of many tumors; however, their effectiveness against triple-negative breast cancers (TNBCs) has been limited. One factor limiting responsiveness of TNBCs to ICIs is a lack of functional tumor-infiltrating lymphocytes (TILs) in 'non-inflamed' or 'cold' tumor immune microenvironments (TIMEs), although by unknown mechanisms. Targeting MUC1-C in a mouse transgenic TNBC tumor model increases cytotoxic tumor-infiltrating CD8+ T cells (CTLs), supporting a role for MUC1-C in immune evasion. The basis for these findings and whether they extend to human TNBCs are not known. METHODS: Human TNBC cells silenced for MUC1-C using short hairpin RNAs (shRNAs) were analyzed for the effects of MUC1-C on global transcriptional profiles. Differential expression and rank order analysis was used for gene set enrichment analysis (GSEA). Gene expression was confirmed by quantitative reverse-transcription PCR and immunoblotting. The The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets were analyzed for effects of MUC1 on GSEA, cell-type enrichment, and tumor immune dysfunction and exclusion. Single-cell scRNA-seq datasets of TNBC samples were analyzed for normalized expression associations between MUC1 and selected genes within tumor cells. RESULTS: Our results demonstrate that MUC1-C is a master regulator of the TNBC transcriptome and that MUC1-C-induced gene expression is driven by STAT1 and IRF1. We found that MUC1-C activates the inflammatory interferon (IFN)-γ-driven JAK1→STAT1→IRF1 pathway and induces the IDO1 and COX2/PTGS2 effectors, which play key roles in immunosuppression. Involvement of MUC1-C in activating the immunosuppressive IFN-γ pathway was extended by analysis of human bulk and scRNA-seq datasets. We further demonstrate that MUC1 associates with the depletion and dysfunction of CD8+ T cells in the TNBC TIME. CONCLUSIONS: These findings demonstrate that MUC1-C integrates activation of the immunosuppressive IFN-γ pathway with depletion of TILs in the TNBC TIME and provide support for MUC1-C as a potential target for improving TNBC treatment alone and in combination with ICIs. Of translational significance, MUC1-C is a druggable target with chimeric antigen receptor (CAR) T cells, antibody-drug conjugates (ADCs) and a functional inhibitor that are under clinical development.


Subject(s)
Gene Expression Profiling/methods , Interferon-gamma/genetics , Mucin-1/genetics , Triple Negative Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Mucin-1/metabolism , STAT1 Transcription Factor/metabolism , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment
14.
Int J Clin Oncol ; 26(4): 744-752, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33387085

ABSTRACT

BACKGROUND: The salvage treatments for biochemical recurrence (BCR) include local external beam radiation therapy (RT) and systemic androgen-deprivation therapy (ADT). METHODS: We reviewed patients who underwent radical prostatectomy (RP) and developed BCR at three institutions. After excluding patients whose nadir prostate-specific antigen (PSA) was higher than 0.2 ng/mL, those who received neoadjuvant/adjuvant therapy, and those whose BCR was not treated until their PSA exceeded 4.0 ng/mL, the remaining 335 patients comprised the cohort of this study. Salvage RT and ADT were performed for 154 and 181 patients, respectively. After the failure of salvage RT, all patients received subsequent ADT. The starting point of this study was the timing of BCR and the endpoint was the development of castration-resistant prostate cancer (CRPC). RESULTS: During the mean follow-up period of 8.5 years after BCR, CRPC was observed in 13 patients administered RT and 24 patients administered ADT. Kaplan-Meier curves demonstrated no significant difference in CRPC-free survival between the RT and ADT groups (10-year CRPC-free survival 89.9 vs. 86.3%, p = 0.199). On the other hand, we found a significant difference in CRPC-free survival between the RT and ADT groups in 50 high-risk patients with two risk factors of Grade Group ≥ 4 and PSA-doubling time < 6 months (10-year CRPC-free survival 73.4 vs. 40.3%, p = 0.040). CONCLUSION: This study revealed that salvage RT increases the CRPC-free survival rate compared with salvage ADT in high-risk patients with Grade Group ≥ 4 and PSA-doubling time < 6 months.


Subject(s)
Androgen Antagonists , Prostatic Neoplasms , Androgen Antagonists/therapeutic use , Androgens , Follow-Up Studies , Humans , Male , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/radiotherapy , Prostate-Specific Antigen , Prostatectomy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/surgery , Retrospective Studies , Salvage Therapy
15.
Cancer Res ; 81(4): 1111-1122, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33323379

ABSTRACT

The Brg/Brahma-associated factor (BAF, mSWI/SNF) chromatin remodeling complex is of importance in development and has been linked to prostate oncogenesis. The oncogenic MUC1-C protein promotes lineage plasticity in the progression of neuroendocrine prostate cancer (NEPC), however, there is no known association between MUC1-C and BAF. We report here that MUC1-C binds directly to the E2F1 transcription factor and that the MUC1-C→E2F1 pathway induces expression of embryonic stem cell-specific BAF (esBAF) components BRG1, ARID1A, BAF60a, BAF155, and BAF170 in castrate-resistant prostate cancer (CRPC) and NEPC cells. In concert with this previously unrecognized pathway, MUC1 was associated with increased expression of E2F1 and esBAF components in NEPC tumors as compared with CRPC, supporting involvement of MUC1-C in activating the E2F1→esBAF pathway with progression to NEPC. MUC1-C formed a nuclear complex with BAF and activated cancer stem cell (CSC) gene signatures and the core pluripotency factor gene network. The MUC1-C→E2F1→BAF pathway was necessary for induction of both the NOTCH1 effector of CSC function and the NANOG pluripotency factor, and collectively, this network drove CSC self-renewal. These findings indicate that MUC1-C promotes NEPC progression by integrating activation of E2F1 and esBAF with induction of NOTCH1, NANOG, and stemness. SIGNIFICANCE: These findings show that MUC1-C, which promotes prostate cancer progression, activates a novel pathway that drives the BAF remodeling complex, induces NOTCH1 and NANOG, and promotes self-renewal of prostate cancer stem cells.


Subject(s)
Carcinoma, Neuroendocrine , Mucin-1/physiology , Multiprotein Complexes/genetics , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cell Self Renewal/genetics , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Multiprotein Complexes/metabolism , Neoplastic Stem Cells/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
16.
Sci Rep ; 10(1): 9193, 2020 Jun 08.
Article in English | MEDLINE | ID: mdl-32513929

ABSTRACT

Topological properties in material science have recently received tremendous attention, as exemplified by topological insulators. As for quantum spin systems, the Haldane chain with integer spins is the best known example1. The realization of a topological quantum phase in the Haldane chain is an innovative research development related to the 2016 Nobel Prize in Physics. For mixed spin chains composed of two different spins, the appearance of a topologically quantized magnetization plateau is predicted by the Lieb-Mattis theorem2 in combination with the Oshikawa-Yamanaka-Affleck criterion3. However, the actual magnetization plateau in the mixed spin chain has not yet been observed. Here, we present a model compound forming a mixed spin-(1/2, 5/2) chain. We observe a clear Lieb-Mattis plateau and well explain it quantitatively. The present results demonstrate a quantum many-body effect based on quantum topology and provide a new stage in the search for topological properties in condensed matter physics.

17.
JCI Insight ; 5(12)2020 06 18.
Article in English | MEDLINE | ID: mdl-32427590

ABSTRACT

Colitis is associated with the development of colorectal cancer (CRC) by largely undefined mechanisms that are critical for understanding the link between inflammation and cancer. Intestinal stem cells (ISCs) marked by leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) expression are of importance in both the inflammatory response to colitis and progression to colitis-associated colon cancer (CACC). Here, we report in human mucin 1-transgenic (MUC1-transgenic) mouse models of CACC, targeting the MUC1-C oncogenic protein suppresses the (a) Lgr5+ ISC population, (b) induction of Myc and core pluripotency stem cell factors, and (c) severity and progression of colitis to dysplasia and cancer. By extension to human colon cancer cells, we demonstrate that MUC1-C drives MYC, forms a complex with MYC on the LGR5 promoter, and activates LGR5 expression. We also show in CRC cells that MUC1-C induces cancer stem cell (CSC) markers (BMI1, ALDH1, FOXA1, LIN28B) and the OCT4, SOX2, and NANOG pluripotency factors. Consistent with conferring the CSC state, targeting MUC1-C suppresses the capacity of CRC cells to promote wound healing, invasion, self-renewal, and tumorigenicity. In analysis of human tissues, MUC1 expression associates with activation of inflammatory pathways, development of colitis, and aggressiveness of CRCs. These results collectively indicate that MUC1-C is of importance for integrating stemness and pluripotency in colitis and CRC. Of clinical relevance, the findings further indicate that MUC1-C represents a potentially previously unrecognized target that is druggable for treating progression of colitis and CRC.


Subject(s)
Carcinogenesis/metabolism , Colorectal Neoplasms/metabolism , Mucin-1/metabolism , Neoplastic Stem Cells/metabolism , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Colonic Neoplasms/metabolism , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mucin-1/genetics , Receptors, G-Protein-Coupled/metabolism
18.
Materials (Basel) ; 13(9)2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32344877

ABSTRACT

Experimental studies into the forced magnetostriction, magnetization, and temperature dependence of permeability in Ni2MnIn and Ni2MnSn ferromagnetic Heusler alloys were performed according to the spin fluctuation theory of itinerant ferromagnetism proposed by Takahashi. We investigated the magnetic field (H) dependence of magnetization (M) at the Curie temperature TC, and at T = 4.2 K, which concerns the ground state of the ferromagnetic state. The M-H result at TC was analyzed by means of the H versus M5 dependence. At 4.2 K, it was investigated by means of an Arrott plot (H/M vs. M2) according to Takahashi's theory. As for Ni2MnIn and Ni2MnSn, the spin fluctuation parameters in k-space (momentum space, TA) and that in energy space (frequency space, T0) obtained at TC and 4.2 K were almost the same. The average values obtained at TC and 4.2 K were TA = 342 K, T0 = 276 K for Ni2MnIn and TA = 447 K, T0 = 279 K for Ni2MnSn, respectively. The forced magnetostriction at TC was also investigated. The forced linear magnetostriction (ΔL/L) and the forced volume magnetostriction (ΔV/V) were proportional to M4, which followed Takahashi's theory. We compared the forced volume magnetostriction ΔV/V and mechanical parameter, bulk modulus K. ΔV/V is inversely proportional to K. We also discuss the spin polarization of Ni2MnIn and other magnetic Heusler alloys. The pC/pS of Ni2MnIn was 0.860. This is comparable with that of Co2MnGa, which is a famous half-metallic alloy.

20.
Nat Commun ; 11(1): 338, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953400

ABSTRACT

Neuroendocrine prostate cancer (NEPC) is an aggressive malignancy with no effective targeted therapies. The oncogenic MUC1-C protein is overexpressed in castration-resistant prostate cancer (CRPC) and NEPC, but its specific role is unknown. Here, we demonstrate that upregulation of MUC1-C in androgen-dependent PC cells suppresses androgen receptor (AR) axis signaling and induces the neural BRN2 transcription factor. MUC1-C activates a MYC→BRN2 pathway in association with induction of MYCN, EZH2 and NE differentiation markers (ASCL1, AURKA and SYP) linked to NEPC progression. Moreover, MUC1-C suppresses the p53 pathway, induces the Yamanaka pluripotency factors (OCT4, SOX2, KLF4 and MYC) and drives stemness. Targeting MUC1-C decreases PC self-renewal capacity and tumorigenicity, suggesting a potential therapeutic approach for CRPC and NEPC. In PC tissues, MUC1 expression associates with suppression of AR signaling and increases in BRN2 expression and NEPC score. These results highlight MUC1-C as a master effector of lineage plasticity driving progression to NEPC.


Subject(s)
Carcinoma, Neuroendocrine/metabolism , Disease Progression , Mucin-1/metabolism , Neuronal Plasticity/physiology , Prostatic Neoplasms/metabolism , Animals , Aurora Kinase A/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinogenesis/genetics , Carcinoma, Neuroendocrine/genetics , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Nude , Mucin-1/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , POU Domain Factors/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Proto-Oncogene Proteins c-myc , SOXB1 Transcription Factors/metabolism , Signal Transduction , Synaptophysin/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...