Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 919
Filter
1.
J Cutan Pathol ; 51(11): 828-833, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38986510

ABSTRACT

PRRX1-fused mesenchymal neoplasm is a recently identified, rare subcutaneous soft tissue neoplasm that is characterized by fusion of PRRX1 (exon 1) with NCOA1 (exon 13) in the majority of reported cases. Although initially considered to be fibroblastic, a possibility of neural or neuroectodermal differentiation has been suggested in a subset of cases. We report a 26-year-old female with a 4.0 cm painless mass located in the subcutis of the left thigh. Microscopically, the tumor was well-circumscribed and multinodular and was composed of relatively monomorphic ovoid to spindle cells arranged in loose fascicles, trabeculae, and cords within alternating myxoid and fibrous matrix, and vascularized stroma. Mitotic figures were scarce and necrosis was not observed. By immunohistochemistry, the neoplastic cells demonstrated focal co-expression of S100 protein and SOX10 and were negative for epithelial membrane antigen, smooth muscle actin, desmin, CD34, STAT6, HMB45, Melan-A, and MUC4. The expression of Rb1 was retained. Targeted RNA-sequencing identified a novel transcript fusion of PRRX1 (exon 1)::NCOA1 (exon 15), which was further confirmed by reverse transcription polymerase chain reaction and Sanger sequencing. The tumor was narrowly excised and no tumor recurrence or metastasis was identified after 13 months of follow-up. In summary, we report a new case of PRRX1-fused mesenchymal neoplasm, expanding the molecular genetic spectrum and providing further support for possible neural or neuroectodermal differentiation of this emerging soft tissue tumor entity.


Subject(s)
Homeodomain Proteins , Nuclear Receptor Coactivator 1 , Soft Tissue Neoplasms , Humans , Female , Adult , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Thigh/pathology
2.
Adv Sci (Weinh) ; 11(33): e2310037, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38953362

ABSTRACT

Programmed death-ligand 1 (PD-L1) is overexpressed in multiple cancers and critical for their immune escape. It has previously shown that the nuclear coactivator SRC-1 promoted colorectal cancer (CRC) progression by enhancing CRC cell viability, yet its role in CRC immune escape is unclear. Here, we demonstrate that SRC-1 is positively correlated with PD-L1 in human CRC specimens. SRC-1 deficiency significantly inhibits PD-L1 expression in CRC cells and retards murine CRC growth in subcutaneous grafts by enhancing CRC immune escape via increasing tumor infiltration of CD8+ T cells. Genetic ablation of SRC-1 in mice also decreases PD-L1 expression in AOM/DSS-induced murine CRC. These results suggest that tumor-derived SRC-1 promotes CRC immune escape by enhancing PD-L1 expression. Mechanistically, SRC-1 activated JAK-STAT signaling by inhibiting SOCS1 expression and coactivated STAT3 and IRF1 to enhance PD-L1 transcription as well as stabilized PD-L1 protein by inhibiting proteasome-dependent degradation mediated by speckle type POZ protein (SPOP). Pharmacological inhibition of SRC-1 improved the antitumor effect of PD-L1 antibody in both subcutaneous graft and AOM/DSS-induced murine CRC models. Taken together, these findings highlight a crucial role of SRC-1 in regulating PD-L1 expression and targeting SRC-1 in combination with PD-L1 antibody immunotherapy may be an attractive strategy for CRC treatment.


Subject(s)
B7-H1 Antigen , Colorectal Neoplasms , Disease Models, Animal , Nuclear Receptor Coactivator 1 , Tumor Escape , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/immunology , Animals , Mice , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Humans , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Tumor Escape/genetics , Tumor Escape/immunology , Protein Stability , Cell Line, Tumor , Mice, Inbred C57BL
3.
Phytomedicine ; 130: 155759, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38788394

ABSTRACT

BACKGROUND: Farnesoid X receptor (FXR) is a vital receptor for bile acids and plays an important role in the treatment of cholestatic liver disease. In addition to traditional bile acid-based steroidal agonists, synthetic alkaloids are the most commonly reported non-steroidal FXR agonists. Sarmentol H is a nor-sesquiterpenoid obtained from Sedum sarmentosum Bunge, and in vitro screening experiments have shown that it might be related to the regulation of the FXR pathway in a previous study. PURPOSE: To investigate the therapeutic effects of sarmentol H on cholestasis and to determine whether sarmentol H directly targets FXR to mitigate cholestasis. Furthermore, this study aimed to explore the key amino acid residues involved in the binding of sarmentol H to FXR through site-directed mutagenesis. METHODS: An intrahepatic cholestasis mouse model was established to investigate the therapeutic effects of sarmentol H on cholestasis. In vitro experiments, including Co-Ip and FXR-EcRE-Luc assays, were performed to assess whether sarmentol H activates FXR by recruiting the receptor coactivator SRC1. CETSA, SIP, DARTS, and ITC were used to determine the binding of sarmentol H to FXR protein. The key amino acid residues for sarmentol H binding to FXR were analyzed by molecular docking and site-directed mutagenesis. Finally, we conducted in vivo experiments on wild-type and Fxr-/- mice to further validate the anticholestatic target of sarmentol H. RESULTS: Sarmentol H had significant ameliorative effects on the pathological conditions of cholestatic mice induced with ANIT. In vitro experiments suggested that it is capable of activating FXR and regulating downstream signaling pathways by recruiting SRC1. The target validation experiments showed that sarmentol H had the ability to bind to FXR as a ligand (KD = 2.55 µmol/L) and enhance the stability of its spatial structure. Moreover, site-directed mutagenesis revealed that THR292 and TYR365 were key binding sites for sarmentol H and FXR. Furthermore, knockout of the Fxr gene resulted in a significantly higher degree of ANIT-induced cholestatic liver injury than that in wild-type cholestatic mice, and the amelioration of cholestasis or regulatory effects on FXR downstream genes by sarmentol H also disappeared in Fxr-/- cholestatic mice. CONCLUSION: Sarmentol H is an FXR agonist. This is the first study to show that it exerts a significant therapeutic effect on cholestatic mice, and can directly bind to FXR and activate it by recruiting the coactivator SRC1.


Subject(s)
Cholestasis , Nuclear Receptor Coactivator 1 , Receptors, Cytoplasmic and Nuclear , Animals , Humans , Male , Mice , Cholestasis/drug therapy , Disease Models, Animal , Hep G2 Cells , Mice, Inbred C57BL , Molecular Docking Simulation , Mutagenesis, Site-Directed , Receptors, Cytoplasmic and Nuclear/metabolism
4.
Int J Biol Macromol ; 268(Pt 1): 131865, 2024 May.
Article in English | MEDLINE | ID: mdl-38670200

ABSTRACT

A previous study reported the use of a biosensing technique based on surface plasmon resonance (SPR) for the ligand binding detection of peroxisome proliferator activator receptor gamma (PPARγ). This detection was designed based on the structural properties of PPARγ. Because of cross-linked protein inactivation and the low molecular weight of conventional ligands, direct ligand binding detection based on SPR has low stability and repeatability. In this study, we report an indirect response methodology based on SPR technology in which anti-His CM5 chip binds fresh PPARγ every cycle, resulting in more stable detection. We developed a remarkable improvement in ligand-protein binding detectability in vitro by introducing two coregulator-related polypeptides into this system. In parallel, a systematic indirect response methodology can reflect the interaction relationship between ligands and proteins to some extent by detecting the changes in SA-SRC1 and GST-NCOR2 binding to PPARγ. Rosiglitazone, a PPARγ agonist with strong affinity, is a potent insulin-sensitizing agent. Some ligands may be competitively exerted at the same sites of PPARγ (binding rosiglitazone). We demonstrated using indirect response methodology that selective PPARγ modulator (SPPARM) candidates of PPARγ can be found by competing for the binding of the rosiglitazone site on PPARγ, although they may have no effect on polypeptides and PPARγ binding.


Subject(s)
Nuclear Receptor Coactivator 1 , PPAR gamma , Protein Binding , Surface Plasmon Resonance , Surface Plasmon Resonance/methods , PPAR gamma/metabolism , PPAR gamma/chemistry , Ligands , Nuclear Receptor Coactivator 1/metabolism , Nuclear Receptor Coactivator 1/chemistry , Peptides/chemistry , Peptides/metabolism , Humans , Rosiglitazone/pharmacology , Nuclear Receptor Co-Repressor 2
5.
J Cell Mol Med ; 28(7): e18171, 2024 04.
Article in English | MEDLINE | ID: mdl-38506084

ABSTRACT

SRC-1 functions as a transcriptional coactivator for steroid receptors and various transcriptional factors. Notably, SRC-1 has been implicated in oncogenic roles in multiple cancers, including breast cancer and prostate cancer. Previous investigations from our laboratory have established the high expression of SRC-1 in human HCC specimens, where it accelerates HCC progression by enhancing Wnt/beta-catenin signalling. In this study, we uncover a previously unknown role of SRC-1 in HCC metastasis. Our findings reveal that SRC-1 promotes HCC metastasis through the augmentation of MMP-9 expression. The knockdown of SRC-1 effectively mitigated HCC cell metastasis both in vitro and in vivo by suppressing MMP-9 expression. Furthermore, we observed a positive correlation between SRC-1 mRNA levels and MMP-9 mRNA levels in limited and larger cohorts of HCC specimens from GEO database. Mechanistically, SRC-1 operates as a coactivator for NF-κB and AP-1, enhancing MMP-9 promoter activity in HCC cells. Higher levels of SRC-1 and MMP-9 expression are associated with worse overall survival in HCC patients. Treatment with Bufalin, known to inhibit SRC-1 expression, significantly decreased MMP-9 expression and inhibited HCC metastasis in both in vitro and in vivo settings. Our results demonstrated the pivotal role of SRC-1 as a critical modulator in HCC metastasis, presenting a potential therapeutic target for HCC intervention.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Male , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , RNA, Messenger , Neoplasm Invasiveness/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic
6.
J Neurooncol ; 163(3): 693-705, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37402091

ABSTRACT

BACKGROUND: Corticosteroid is commonly used before surgery to control cerebral oedema in brain tumours and is frequently continued throughout treatment. Its long-term effect of on the recurrence of WHO-Grade 4 astrocytoma remains controversial. The interaction between corticosteroid, SRC-1 gene and cytotoxic T-cells has never been investigated. METHODS: A retrospective cohort of 36 patients with WHO-Grade 4 astrocytoma were examined for CD8 + T-cell and SRC-1 gene expressions through IHC and qRT-PCR. The impact of corticosteroid on CD8+T-cells infiltration, SRC-1 expression, and tumour recurrence was analyzed. RESULTS: The mean patients age was 47-years, with a male to female ratio 1.2. About 78% [n = 28] of the cases showed reduced or no CD8+T-cell expression while 22% [n = 8] of cases have showed medium to high CD8+T-cell expression. SRC-1 gene was upregulated in 5 cases [14%] and 31 cases [86%] showed SRC-1 downregulation. The average of total days and doses of administered corticosteroid from the preoperative period to the postoperative period was at range of 14-106 days and 41-5028 mg, respectively. There was no significant statistical difference in RFI among tumours expressing high or low CD8+T-cells when corticosteroid was administered in recommended or exceeded doses [p-value = 0.640]. There was a significant statistical difference in RFI between CD8+T-Cell expression and SRC-1 gene dysregulation [p-value = 002]. Tumours with high CD8+T T-cell expression and SRC-1 gene downregulation had late recurrence. CONCLUSIONS: Corticosteroid treatment can directly affect the SRC-1 gene regulation but does not directly influence cytotoxic T-cells infiltration or tumor progression. However, SRC-1 gene downregulation can facilitate late tumor recurrence.


Subject(s)
Astrocytoma , Glioblastoma , Nuclear Receptor Coactivator 1 , Female , Humans , Male , Middle Aged , Adrenal Cortex Hormones/therapeutic use , Astrocytoma/drug therapy , Astrocytoma/genetics , Astrocytoma/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Retrospective Studies , World Health Organization , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism
7.
Life Sci ; 320: 121555, 2023 May 01.
Article in English | MEDLINE | ID: mdl-36878279

ABSTRACT

BACKGROUND: Matrix metallopeptidases (MMPs) are critical matrix-degrading molecules and they are frequently overexpressed in degenerative discs. This study aimed to investigate the mechanism for MMP upregulation. METHODS: Immunoblot and RT-qPCR were used for detecting protein and gene expression levels. 4-month-old and 24-month-old C57BL/6 mice were used for evaluating intervertebral disc degeneration (IDD). An ubiquitination assay was used to determine protein modification. Immunoprecipitation and mass spectrometry were used for identifying protein complex members. RESULTS: We identified the elevation of 14 MMPs among 23 members in aged mice with IDD. Eleven of these 14 MMP gene promoters contained a Runx2 (runt-related transcription factor 2) binding site. Biochemical analyses revealed that Runx2 recruited a histone acetyltransferase p300 and a coactivator NCOA1 (nuclear receptor coactivator 1) to assemble a complex, transactivating MMP expression. The deficiency of an E3 ligase called HERC3 (HECT and RLD domain containing E3 ubiquitin-protein ligase 3) resulted in the accumulation of NCOA1 in the inflammatory microenvironment. High throughput screening of small molecules that specifically target the NCOA1-p300 interaction identified a compound SMTNP-191, which showed an inhibitory effect on suppressing MMP expression and attenuating the IDD process in aged mice. CONCLUSION: Our data support a model in which deficiency of HERC3 fails to ubiquitinate NCOA1, leading to the assembly of NCOA1-p300-Runx2 and causing the transactivation of MMPs. These findings offer new insight into inflammation-mediated MMP accumulation and also provide a new therapeutic strategy to retard the IDD process.


Subject(s)
Intervertebral Disc Degeneration , Intervertebral Disc , Mice , Animals , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Nuclear Receptor Coactivator 1 , Mice, Inbred C57BL , Extracellular Matrix/metabolism , Metalloproteases/metabolism , Intervertebral Disc/metabolism
9.
Cytokine ; 165: 156186, 2023 05.
Article in English | MEDLINE | ID: mdl-36989655

ABSTRACT

Substance P (SP), a neuropeptide consisting of 11 amino acid residues, is involved in the pathogenesis of encephalomyocarditis virus (EMCV)-induced myocarditis by stimulating the production of proinflammatory cytokines. However, the underlying mechanism that regulates SP production is still unknown. In this study, we report the transcriptional regulation of the Tachykinin Precursor 1 (TAC1) gene that encodes SP by a transcriptional complex composed of Steroid Receptor Coactivator 1 (Src1), Peroxisome proliferator-activated receptor-gamma coactivator 1 (PGC1α), and Activator Protein 1 (AP1) transcription factor. Infection of mice with EMCV induced the accumulation of PGC1α and increased TAC1 expression, thereby promoting the secretion of SP, initiating apoptosis, and elevating proinflammatory cytokine levels. In vitro overexpression of the Src1-PGC1α-AP1 members also induced TAC1 expression, increased the SP concentration, initiated apoptosis, and elevated proinflammatory cytokine concentrations. Depletion or inhibition of the Src1-PGC1α-AP1 complex reversed these effects. The administration of gossypol, an Src1 inhibitor, or SR1892, a PGC1α inhibitor, to EMCV-infected mice attenuated myocarditis. Taken together, our results reveal that the upregulation of TAC1 and the secretion of SP in EMCV-induced myocarditis are dependent on the Src1-PGC1α-AP1 complex. Targeting the Src1-PGC1α-AP1 complex may represent a new therapeutic strategy for myocarditis.


Subject(s)
Encephalomyocarditis virus , Myocarditis , Animals , Mice , Apoptosis , Cytokines/metabolism , Encephalomyocarditis virus/metabolism , Inflammation , Myocarditis/metabolism , Nuclear Receptor Coactivator 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Substance P , Transcription Factor AP-1/metabolism
11.
Hum Pathol ; 135: 65-75, 2023 05.
Article in English | MEDLINE | ID: mdl-36646185

ABSTRACT

Recurrent NCOA1/2/3 gene fusions emerged in uterine tumor resembling ovarian sex cord tumor (UTROSCT). More cases are required to consolidate these molecular alterations. In this study, the clinicopathological features and immunostaining profiles were reviewed in 18 UTROSCT. Fluorescence in situ hybridization for dual color break-apart probes of NCOA1, NCOA2, NCOA3, BCOR, YWHAE, PHF1 and JAZF1 were performed on 16 tumors. Eight cases were subjected to targeted next-generation sequencing to detect genomic alterations. We found that the tumors predominantly showed various sex-cord patterns without a recognizable endometrial stromal component. They exhibited a diverse immunohistochemical profile, frequently co-expressing sex cord (calretinin, inhibin, WT1, SF-1, and FOXL2), smooth muscle (SMA, desmin and caldesmon), epithelial (CK) and other markers (CD10 and IFITM1). Fourteen of 16 tumors (87.5%) showed NCOA1-3 gene rearranges, but none had BCOR, YWHAE, PHF1 and JAZF1 fusions. Five tumors contained 6 non-recurrent pathogenic (likely) mutations and one had gains in c-MYC. Our study supports frequent NCOA1-3 rearrangements in UTROSCT. Rare, non-recurrent mutations suggest that these gene rearrangements be potential drivers in tumorigenesis. Detection of gene rearrangements can contribute to the correct interpretation of UTROSCT. However, large comparative studies with molecular tests are required to confirm these findings.


Subject(s)
Ovarian Neoplasms , Sex Cord-Gonadal Stromal Tumors , Uterine Neoplasms , Female , Humans , In Situ Hybridization, Fluorescence , Uterine Neoplasms/pathology , Sex Cord-Gonadal Stromal Tumors/diagnosis , Transcription Factors/genetics , Gene Rearrangement , Biomarkers, Tumor/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Nuclear Receptor Coactivator 1/genetics
12.
Int J Surg Pathol ; 31(2): 227-232, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35477326

ABSTRACT

MEIS1::NCOA1/2 sarcomas are a newly recognized group of exceedingly rare low-grade spindle cell sarcomas that often involve the genitourinary and gynecologic tracts. Due to its deceptively low-grade morphology and the non-specific immunoprofile, these neoplasms may pose a diagnostic challenge by histologically mimicking other entities such as endometrial stromal sarcoma, smooth muscle tumor, or uterine perivascular epithelioid cell tumor (PEComa). Histologically, MEIS1::NCOA1/2 sarcomas typically show spindle cell proliferation with hyperchromatic nuclei and a generalized cytologic uniformity, arranged in short fascicles and exhibiting alternating zones of hypo- and hypercellularity. Among the previously reported cases, molecular analysis revealed the MEIS1::NCOA2 fusion as the most commonly detected fusion gene, whereas the MEIS1::NCOA1 fusion gene has been reported in only a single case that involved kidney. Herein we report the first case of uterine sarcoma harboring the MEIS1::NCOA1 fusion gene that was initially misclassified as low-grade endometrial stromal sarcoma, demonstrating its clinicopathologic features, and highlighting the essential role of molecular pathology to arrive at the accurate diagnosis that may alter disease classification and inform therapy.


Subject(s)
Endometrial Neoplasms , Sarcoma, Endometrial Stromal , Uterine Neoplasms , Humans , Female , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Sarcoma, Endometrial Stromal/diagnosis , Sarcoma, Endometrial Stromal/genetics , Sarcoma, Endometrial Stromal/pathology , Uterine Neoplasms/diagnosis , Uterine Neoplasms/genetics , Uterus/pathology , Nuclear Receptor Coactivator 1/genetics
13.
J Mol Biol ; 434(24): 167869, 2022 12 30.
Article in English | MEDLINE | ID: mdl-36309135

ABSTRACT

Steroid receptors (SRs) are ligand-dependent transcription factors (TFs) relevant to key cellular processes in both physiology and pathology, including some types of cancer. SOX2 is a master TF of pluripotency and self-renewal of embryonic stem cells, and its dysregulation is also associated with various types of human cancers. A potential crosstalk between these TFs could be relevant in malignant cells yet, to the best of our knowledge, no formal study has been performed thus far. Here we show, by quantitative live-cell imaging microscopy, that ectopic expression of SOX2 disrupts the formation of hormone-dependent intranuclear condensates of many steroid receptors (SRs), including those formed by the glucocorticoid receptor (GR). SOX2 also reduces GR's binding to specific DNA targets and modulates its transcriptional activity. SOX2-driven effects on GR condensates do not require the intrinsically disordered N-terminal domain of the receptor and, surprisingly, neither relies on GR/SOX2 interactions. SOX2 also alters the intranuclear dynamics and compartmentalization of the SR coactivator NCoA-2 and impairs GR/NCoA-2 interactions. These results suggest an indirect mechanism underlying SOX2-driven effects on SRs involving this coactivator. Together, these results highlight that the transcriptional program elicited by GR relies on its nuclear organization and is intimately linked to the distribution of other GR partners, such as the NCoA-2 coactivator. Abnormal expression of SOX2, commonly observed in many tumors, may alter the biological action of GR and, probably, other SRs as well. Understanding this crosstalk may help to improve steroid hormone-based therapies in cancers with elevated SOX2 expression.


Subject(s)
Receptors, Glucocorticoid , SOXB1 Transcription Factors , Transcriptional Activation , Humans , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
14.
Life Sci ; 307: 120906, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36007610

ABSTRACT

Targeted therapy is receiving considerable attention from the researchers around the globe owing to the increased drug-resistance and incidences of cancer recurrences. MicroRNAs (miRNAs) exhibits tremendous potential as a candidate for molecular targeted therapy in cancer. Unfortunately, majority of research related to microRNAs are focussed on either a particular miRNA or a set of unrelated miRNAs. There is lack of holistic knowledge on differential co-expression of miRNA clusters in regulating the gene expression under physiological conditions. Previously, we reported the cooperative effect of hsa-miR-23a~27a~24-2 cluster in inducing ER (Endoplasmic Reticulum) stress-mediated apoptotic cell death of HEK cells. In the present study, we have investigated the common anti-cancer effects of individual members of this cluster. Our in silico analysis identified twelve common target genes distributed across three independent clusters. Furthermore, we found NCOA1, NLK, and RAP1B to fall in a single cluster with NCOA1 as a central hub molecule. Prognostic analysis showed profound involvement of these three genes in the breast cancer progression and metastasis. We further demonstrated that alteration in the levels of individual members of miR-23a~27a~24-2 cluster commonly regulates the invasive migration of breast cancer cells by modulating EMT and cytoskeletal pathway proteins. Our results reveal a new insight into the therapeutic potential of individual members of the pro-apoptotic hsa-miR-23a~27a~24-2 cluster family against metastatic breast cancer.


Subject(s)
Breast Neoplasms , MicroRNAs , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Endoplasmic Reticulum Stress , Female , Humans , MicroRNAs/metabolism , Neoplasm Recurrence, Local , Nuclear Receptor Coactivator 1 , Protein Serine-Threonine Kinases , rap GTP-Binding Proteins/metabolism
15.
J Mol Biol ; 434(16): 167718, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35810793

ABSTRACT

Nuclear receptors constitute one of the largest families of transcription factors that regulate genes in metazoans in response to small molecule ligands. Many receptors harbor two transactivation domains, one at each end of the protein sequence. Whereas the molecular mechanisms of transactivation mediated by the ligand-binding domain at the C-terminus of the protein are generally well established, the mechanism involving the N-terminal domain called activation function 1 (AF1) has remained elusive. Previous studies implicated the AF1 domain as a significant contributor towards the overall transcriptional activity of the NR4A family of nuclear receptors and suggested that the steroid receptor coactivators (SRCs) play an important role in this process. Here we show that a short segment within the AF1 domain of the NR4A receptor Nurr1 can directly engage with the SRC1 PAS-B domain. We also show that this segment forms a helix upon binding to a largely hydrophobic groove on PAS-B, overlapping with the surface engaged by the STAT6 transcription factor, suggesting that this mode of recruitment could be shared by diverse transcription factors including other nuclear receptors.


Subject(s)
Nuclear Receptor Coactivator 1 , Nuclear Receptor Subfamily 4, Group A, Member 2 , Transcriptional Activation , Amino Acid Sequence , Nuclear Receptor Coactivator 1/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 2/chemistry , Protein Binding , Protein Domains , STAT6 Transcription Factor/chemistry
16.
Crit Rev Eukaryot Gene Expr ; 32(4): 21-29, 2022.
Article in English | MEDLINE | ID: mdl-35695662

ABSTRACT

The regulatory mechanism and function of steroid receptor coactivator-1 (SRC-1) was determined in vitro and the role played in gastric cancer was investigated. The study collected 64 patients with gastric cancer tissue and paracancerous tissue to investigate the clinical patterns of SRC-1 expression in gastric cancer. Quantitative polymerase chain reaction, Western blot, enzyme-linked immunosorbent assay, and immunofluorescence staining were used in this study. In patients with gastric cancer, SRC-1 serum expression levels were up-regulated. Over-expression of SRC-1 promoted cell growth and cell metastasis in vitro model of gastric cancer. However, down-regulation of SRC-1 reduced cell growth and cell metastasis in vitro model of gastric cancer. SRC-1 over-expression induced vascular endothelial growth factor C (VEGFC) protein expressions in vitro model by activation of nuclear factor-kappa B (NF-kB) expression. The inhibition of NF-κB reduced the pro-cancer effects of SRC-1 on cell growth and cell metastasis in vitro model of gastric cancer through inhibition of VEGFC expression. These results suggest that SRC-1 promoted cell metastasis of gastric cancer via VEGFC activator by NF-κB. These novel findings may shed further light on the pathogenesis of gastric cancer and on potential precursor markers.


Subject(s)
Nuclear Receptor Coactivator 1/blood , Receptors, Steroid , Stomach Neoplasms , Cell Line, Tumor , Humans , NF-kappa B/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism
17.
J Cutan Pathol ; 49(9): 802-807, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35583270

ABSTRACT

PRRX::NCOAx-rearranged fibroblastic tumor is a recently described, morphologically distinctive subcutaneous fibroblastic tumor with benign behavior. To date, 12 cases have been reported. Here, we report a new case of PRRX::NCOAx-rearranged fibroblastic tumor showing a prominent pigmented component. The lesion occurred on the shoulder of a 23-year-old male. It was an at least 2.5 cm subcutaneous tumor with a multinodular and plexiform appearance. Morphologically, the tumor was characterized by a variably cellular proliferation of uniform oval to spindle cells arranged in fascicles and cords within a myxocollagenous stroma. Irregular, elongated, dilated vessels were prominent at the periphery of tumor nodules. In addition, nests and clusters of pigment-laden epithelioid and dendritic cells were present. Immunohistochemically, the non-pigmented tumor cells showed patchy positivity for factor XIIIa and focal positivity for S100 protein. The pigmented cells were positive for S100 protein, SOX10, MITF, and a pan-melanocytic cocktail (Melan-A, HMB-45, and tyrosinase). Next-generation RNA sequencing identified an in-frame PRRX1::NCOA1 fusion. In summary, this case highlights a rare pigmented variant of PRRX::NCOAx-rearranged fibroblastic tumor, expanding the morphologic spectrum of this newly described mesenchymal tumor.


Subject(s)
Biomarkers, Tumor , Neoplasms, Fibrous Tissue , Adult , Biomarkers, Tumor/genetics , Gene Fusion , Homeodomain Proteins/genetics , Humans , Male , Nuclear Receptor Coactivator 1/genetics , S100 Proteins/genetics , SOXE Transcription Factors/genetics , Young Adult
18.
Oncogene ; 41(20): 2846-2859, 2022 05.
Article in English | MEDLINE | ID: mdl-35418691

ABSTRACT

Overexpression of nuclear coactivator steroid receptor coactivator 1 (SRC-1) and aberrant activation of the Hedgehog (Hh) signaling pathway are associated with various tumorigenesis; however, the significance of SRC-1 in colorectal cancer (CRC) and its contribution to the activation of Hh signaling are unclear. Here, we identified a conserved Hh signaling signature positively correlated with SRC-1 expression in CRC based on TCGA database; SRC-1 deficiency significantly inhibited the proliferation, survival, migration, invasion, and tumorigenesis of both human and mouse CRC cells, and SRC-1 knockout significantly suppressed azoxymethane/dextran sodium sulfate (AOM/DSS)-induced CRC in mice. Mechanistically, SRC-1 promoted the expression of GLI family zinc finger 2 (GLI2), a major downstream transcription factor of Hh pathway, and cooperated with GLI2 to enhance multiple Hh-regulated oncogene expression, including Cyclin D1, Bcl-2, and Slug. Pharmacological blockages of SRC-1 and Hh signaling retarded CRC progression in human CRC cell xenograft mouse model. Together, our studies uncover an SRC-1/GLI2-regulated Hh signaling looping axis that promotes CRC tumorigenesis, offering an attractive strategy for CRC treatment.


Subject(s)
Colorectal Neoplasms , Hedgehog Proteins , Nuclear Receptor Coactivator 1 , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Humans , Mice , Nuclear Proteins/genetics , Nuclear Receptor Coactivator 1/genetics , Signal Transduction/physiology , Zinc Finger Protein Gli2/metabolism
19.
Exp Cell Res ; 415(2): 113114, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35339471

ABSTRACT

Inflammatory pathways represented by TLR4/NF-κB (Toll-like receptor 4/Nuclear factor-κB) axis signaling are activated in the pathogenesis of endotoxin-induced myocardial dysfunction (EIMD). However, the underlying mechanism by which NF-κB coordinates with other transcriptional coactivators/corepressors to regulate the expression of proinflammatory cytokine genes remains unclear. We established an EIMD-mouse model by intraperitoneal injection of lipopolysaccharides (LPS), and we discovered that NCOA1 (nuclear receptor coactivator 1) assembled with CBP (CREB binding protein) and NF-κB subunits to form a transcriptional complex that specifically bound to promoters of proinflammatory cytokine genes to activate their expression. LPS treatment also inhibited DNMT1 (DNA methyltransferase 1) expression, thereby decreasing DNA methylation of a CpG island located on the promoter of NCOA1 and causing NCOA1 overexpression. Screening small molecules that abolished NCOA1-CBP interaction in a yeast system identified a compound PSSM2126 that effectively blocked the NCOA1-CBP interaction in vitro and in vivo. Administration of PSSM2126 to EIMD mice significantly alleviated the inflammation response and improved cardiac function. Collectively, our results reveal that an NCOA1-dependent transactivation mechanism can regulate proinflammatory cytokine expression, thereby improving our understanding of the activation of NF-κB targets. The promising inhibition of the NCOA1-CBP interaction by PSSM2126 may provide a new therapeutic option for EIMD.


Subject(s)
CREB-Binding Protein , Heart , Inflammation , NF-kappa B , Nuclear Receptor Coactivator 1 , Animals , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cytokines/metabolism , Endotoxins , Heart/physiopathology , Inflammation/chemically induced , Inflammation/genetics , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism
20.
J Clin Endocrinol Metab ; 107(6): e2532-e2544, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35137184

ABSTRACT

CONTEXT: Genetic variants affecting the nuclear hormone receptor coactivator steroid receptor coactivator, SRC-1, have been identified in people with severe obesity and impair melanocortin signaling in cells and mice. As a result, obese patients with SRC-1 deficiency are being treated with a melanocortin 4 receptor agonist in clinical trials. OBJECTIVE: Here, our aim was to comprehensively describe and characterize the clinical phenotype of SRC-1 variant carriers to facilitate diagnosis and clinical management. METHODS: In genetic studies of 2462 people with severe obesity, we identified 23 rare heterozygous variants in SRC-1. We studied 29 adults and 18 children who were SRC-1 variant carriers and performed measurements of metabolic and endocrine function, liver imaging, and adipose tissue biopsies. Findings in adult SRC-1 variant carriers were compared to 30 age- and body mass index (BMI)-matched controls. RESULTS: The clinical spectrum of SRC-1 variant carriers included increased food intake in children, normal basal metabolic rate, multiple fractures with minimal trauma (40%), persistent diarrhea, partial thyroid hormone resistance, and menorrhagia. Compared to age-, sex-, and BMI-matched controls, adult SRC-1 variant carriers had more severe adipose tissue fibrosis (46.2% vs 7.1% respectively, P = .03) and a suggestion of increased liver fibrosis (5/13 cases vs 2/13 in controls, odds ratio = 3.4), although this was not statistically significant. CONCLUSION: SRC-1 variant carriers exhibit hyperphagia in childhood, severe obesity, and clinical features of partial hormone resistance. The presence of adipose tissue fibrosis and hepatic fibrosis in young patients suggests that close monitoring for the early development of obesity-associated metabolic complications is warranted.


Subject(s)
Nuclear Receptor Coactivator 1 , Obesity, Morbid , Female , Fibrosis , Humans , Male , Nuclear Receptor Coactivator 1/genetics , Obesity, Morbid/complications , Obesity, Morbid/genetics
SELECTION OF CITATIONS
SEARCH DETAIL