Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 727
Filter
Add more filters

Publication year range
1.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38657972

ABSTRACT

Advances in fluorescence microscopy and tissue-clearing have revolutionised 3D imaging of fluorescently labelled tissues, organs and embryos. However, the complexity and high cost of existing software and computing solutions limit their widespread adoption, especially by researchers with limited resources. Here, we present Acto3D, an open-source software, designed to streamline the generation and analysis of high-resolution 3D images of targets labelled with multiple fluorescent probes. Acto3D provides an intuitive interface for easy 3D data import and visualisation. Although Acto3D offers straightforward 3D viewing, it performs all computations explicitly, giving users detailed control over the displayed images. Leveraging an integrated graphics processing unit, Acto3D deploys all pixel data to system memory, reducing visualisation latency. This approach facilitates accurate image reconstruction and efficient data processing in 3D, eliminating the need for expensive high-performance computers and dedicated graphics processing units. We have also introduced a method for efficiently extracting lumen structures in 3D. We have validated Acto3D by imaging mouse embryonic structures and by performing 3D reconstruction of pharyngeal arch arteries while preserving fluorescence information. Acto3D is a cost-effective and efficient platform for biological research.


Subject(s)
Imaging, Three-Dimensional , Software , Imaging, Three-Dimensional/methods , Animals , Mice , Microscopy, Fluorescence/methods , Optical Imaging/methods , Image Processing, Computer-Assisted/methods , Embryo, Mammalian/diagnostic imaging
2.
Nucleic Acids Res ; 52(1): 141-153, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-37985205

ABSTRACT

Genetic modification of specific genes is emerging as a useful tool to enhance the functions of antitumor T cells in adoptive immunotherapy. Current advances in CRISPR/Cas9 technology enable gene knockout during in vitro preparation of infused T-cell products through transient transfection of a Cas9-guide RNA (gRNA) ribonucleoprotein complex. However, selecting optimal gRNAs remains a major challenge for efficient gene ablation. Although multiple in silico tools to predict the targeting efficiency have been developed, their performance has not been validated in cultured human T cells. Here, we explored a strategy to select optimal gRNAs using our pooled data on CRISPR/Cas9-mediated gene knockout in human T cells. The currently available prediction tools alone were insufficient to accurately predict the indel percentage in T cells. We used data on the epigenetic profiles of cultured T cells obtained from transposase-accessible chromatin with high-throughput sequencing (ATAC-seq). Combining the epigenetic information with sequence-based prediction tools significantly improved the gene-editing efficiency. We further demonstrate that epigenetically closed regions can be targeted by designing two gRNAs in adjacent regions. Finally, we demonstrate that the gene-editing efficiency of unstimulated T cells can be enhanced through pretreatment with IL-7. These findings enable more efficient gene editing in human T cells.


Subject(s)
CRISPR-Cas Systems , Gene Knockout Techniques , T-Lymphocytes , Humans , CRISPR-Cas Systems/genetics , Gene Editing , T-Lymphocytes/metabolism
3.
Int Immunol ; 36(7): 353-364, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38517027

ABSTRACT

The efficient generation of chimeric antigen receptor (CAR) T cells is highly influenced by the quality of apheresed T cells. Healthy donor-derived T cells usually proliferate better than patients-derived T cells and are precious resources to generate off-the-shelf CAR-T cells. However, relatively little is known about the determinants that affect the efficient generation of CAR-T cells from healthy donor-derived peripheral blood mononuclear cells (PBMCs) compared with those from the patients' own PBMCs. We here examined the efficiency of CAR-T cell generation from multiple healthy donor samples and analyzed its association with the phenotypic features of the starting peripheral blood T cells. We found that CD62L expression levels within CD8+ T cells were significantly correlated with CAR-T cell expansion. Moreover, high CD62L expression within naïve T cells was associated with the efficient expansion of T cells with a stem cell-like memory phenotype, an indicator of high-quality infusion products. Intriguingly, genetic disruption of CD62L significantly impaired CAR-T cell proliferation and cytokine production upon antigen stimulation. Conversely, ectopic expression of a shedding-resistant CD62L mutant augmented CAR-T cell effector functions compared to unmodified CAR-T cells, resulting in improved antitumor activity in vivo. Collectively, we identified the surface expression of CD62L as a concise indicator of potent T-cell proliferation. CD62L expression is also associated with the functional properties of CAR-T cells. These findings are potentially applicable to selecting optimal donors to massively generate CAR-T cell products.


Subject(s)
Immunotherapy, Adoptive , L-Selectin , Receptors, Chimeric Antigen , L-Selectin/metabolism , L-Selectin/immunology , Humans , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Animals , Mice , Immunotherapy, Adoptive/methods , Cell Proliferation
4.
J Virol ; 97(5): e0043823, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37042780

ABSTRACT

Viral protein assembly and virion budding are tightly regulated to enable the proper formation of progeny virions. At this late stage in the virus life cycle, some enveloped viruses take advantage of the host endosomal sorting complex required for transport (ESCRT) machinery, which contributes to the physiological functions of membrane modulation and abscission. Bullet-shaped viral particles are unique morphological characteristics of rhabdoviruses; however, the involvement of host factors in rhabdovirus infection and, specifically, the molecular mechanisms underlying virion formation are not fully understood. In the present study, we used a small interfering RNA (siRNA) screening approach and found that the ESCRT-I component TSG101 contributes to the propagation of rabies virus (RABV). We demonstrated that the matrix protein (M) of RABV interacts with TSG101 via the late domain containing the PY and YL motifs, which are conserved in various viral proteins. Loss of the YL motif in the RABV M or the downregulation of host TSG101 expression resulted in the intracellular aggregation of viral proteins and abnormal virus particle formation, indicating a defect in the RABV assembly and budding processes. These results indicate that the interaction of the RABV M and TSG101 is pivotal for not only the efficient budding of progeny RABV from infected cells but also for the bullet-shaped virion morphology. IMPORTANCE Enveloped viruses bud from cells with the host lipid bilayer. Generally, the membrane modulation and abscission are mediated by host ESCRT complexes. Some enveloped viruses utilize their late (L-) domain to interact with ESCRTs, which promotes viral budding. Rhabdoviruses form characteristic bullet-shaped enveloped virions, but the underlying molecular mechanisms involved remain elusive. Here, we showed that TSG101, one of the ESCRT components, supports rabies virus (RABV) budding and proliferation. TSG101 interacted with RABV matrix protein via the L-domain, and the absence of this interaction resulted in intracellular virion accumulation and distortion of the morphology of progeny virions. Our study reveals that virion formation of RABV is highly regulated by TSG101 and the virus matrix protein.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Rabies virus , Rabies , Humans , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Morphogenesis , Rabies/metabolism , Rabies virus/genetics , Rabies virus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/metabolism , Virus Release , Cell Line , Animals
5.
Blood ; 139(14): 2156-2172, 2022 04 07.
Article in English | MEDLINE | ID: mdl-34861037

ABSTRACT

Adoptive cancer immunotherapy can induce objective clinical efficacy in patients with advanced cancer; however, a sustained response is achieved in a minority of cases. The persistence of infused T cells is an essential determinant of a durable therapeutic response. Antitumor T cells undergo a genome-wide remodeling of the epigenetic architecture upon repeated antigen encounters, which inevitably induces progressive T-cell differentiation and the loss of longevity. In this study, we identified PR domain zinc finger protein 1 (PRDM1) ie, Blimp-1, as a key epigenetic gene associated with terminal T-cell differentiation. The genetic knockout of PRDM1 by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) supported the maintenance of an early memory phenotype and polyfunctional cytokine secretion in repeatedly stimulated chimeric antigen receptor (CAR)-engineered T cells. PRDM1 disruption promoted the expansion of less differentiated memory CAR-T cells in vivo, which enhanced T-cell persistence and improved therapeutic efficacy in multiple tumor models. Mechanistically, PRDM1-ablated T cells displayed enhanced chromatin accessibility of the genes that regulate memory formation, thereby leading to the acquisition of gene expression profiles representative of early memory T cells. PRDM1 knockout also facilitated maintaining an early memory phenotype and cytokine polyfunctionality in T-cell receptor-engineered T cells as well as tumor-infiltrating lymphocytes. In other words, targeting PRDM1 enabled the generation of superior antitumor T cells, which is potentially applicable to a wide range of adoptive cancer immunotherapies.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Cytokines , Gene Knockout Techniques , Humans , Lymphocyte Activation , Positive Regulatory Domain I-Binding Factor 1/genetics
6.
BMC Cancer ; 24(1): 554, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698344

ABSTRACT

BACKGROUND: Prostate cancer is dependent on androgen receptor (AR) signaling, and androgen deprivation therapy (ADT) has proven effective in targeting prostate cancer. However, castration-resistant prostate cancer (CRPC) eventually emerges. AR signaling inhibitors (ARSI) have been also used, but resistance to these agents develops due to genetic AR alterations and epigenetic dysregulation. METHODS: In this study, we investigated the role of OCT1, a member of the OCT family, in an AR-positive CRPC patient-derived xenograft established from a patient with resistance to ARSI and chemotherapy. We conducted a genome-wide analysis chromatin immunoprecipitation followed by sequencing and bioinformatic analyses using public database. RESULTS: Genome-wide analysis of OCT1 target genes in PDX 201.1 A revealed distinct OCT1 binding sites compared to treatment-naïve cells. Bioinformatic analyses revealed that OCT1-regulated genes were associated with cell migration and immune system regulation. In particular, C-terminal Binding Protein 2 (CTBP2), an OCT1/AR target gene, was correlated with poor prognosis and immunosuppressive effects in the tumor microenvironment. Metascape revealed that CTBP2 knockdown affects genes related to the immune response to bacteria. Furthermore, TISIDB analysis suggested the relationship between CTBP2 expression and immune cell infiltration in prostate cancer, suggesting that it may contribute to immune evasion in CRPC. CONCLUSIONS: Our findings shed light on the genome-wide network of OCT1 and AR in AR-positive CRPC and highlight the potential role of CTBP2 in immune response and tumor progression. Targeting CTBP2 may represent a promising therapeutic approach for aggressive AR-positive CRPC. Further validation will be required to explore novel therapeutic strategies for CRPC management.


Subject(s)
Alcohol Oxidoreductases , Co-Repressor Proteins , Gene Expression Regulation, Neoplastic , Octamer Transcription Factor-1 , Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Male , Humans , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Mice , Animals , Octamer Transcription Factor-1/metabolism , Octamer Transcription Factor-1/genetics , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Up-Regulation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Xenograft Model Antitumor Assays , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Tumor Microenvironment , Signal Transduction
7.
RNA Biol ; 21(1): 1-17, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38551131

ABSTRACT

RNA-binding proteins (RBPs) play crucial roles in the functions and homoeostasis of various tissues by regulating multiple events of RNA processing including RNA splicing, intracellular RNA transport, and mRNA translation. The Drosophila behavior and human splicing (DBHS) family proteins including PSF/SFPQ, NONO, and PSPC1 are ubiquitously expressed RBPs that contribute to the physiology of several tissues. In mammals, DBHS proteins have been reported to contribute to neurological diseases and play crucial roles in cancers, such as prostate, breast, and liver cancers, by regulating cancer-specific gene expression. Notably, in recent years, multiple small molecules targeting DBHS family proteins have been developed for application as cancer therapeutics. This review provides a recent overview of the functions of DBHS family in physiology and pathophysiology, and discusses the application of DBHS family proteins as promising diagnostic and therapeutic targets for cancers.


Subject(s)
Drosophila , Neoplasms , Male , Animals , Humans , Drosophila/genetics , Drosophila/metabolism , RNA-Binding Proteins/metabolism , RNA Splicing , RNA/metabolism , Neoplasms/genetics , PTB-Associated Splicing Factor/metabolism , Mammals/genetics
8.
Mol Cell ; 64(1): 176-188, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27716482

ABSTRACT

How deregulation of chromatin modifiers causes malignancies is of general interest. Here, we show that histone H2A T120 is phosphorylated in human cancer cell lines and demonstrate that this phosphorylation is catalyzed by hVRK1. Cyclin D1 was one of ten genes downregulated upon VRK1 knockdown in two different cell lines and showed loss of H2A T120 phosphorylation and increased H2A K119 ubiquitylation of its promoter region, resulting in impaired cell growth. In vitro, H2A T120 phosphorylation and H2A K119 ubiquitylation are mutually inhibitory, suggesting that histone phosphorylation indirectly activates chromatin. Furthermore, expression of a phosphomimetic H2A T120D increased H3 K4 methylation. Finally, both VRK1 and the H2A T120D mutant histone transformed NIH/3T3 cells. These results suggest that histone H2A T120 phosphorylation by hVRK1 causes inappropriate gene expression, including upregulated cyclin D1, which promotes oncogenic transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Histones/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromatin/chemistry , Chromatin/metabolism , Cyclin D1/metabolism , Drosophila Proteins , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , HeLa Cells , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Methylation , Mice , Oligopeptides/genetics , Oligopeptides/metabolism , Phosphorylation , Protamine Kinase/genetics , Protamine Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Threonine/metabolism , Ubiquitination
9.
Am J Emerg Med ; 78: 102-111, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38244243

ABSTRACT

INTRODUCTION: The widespread incorporation of extracorporeal cardiopulmonary resuscitation (ECPR) for out-of-hospital cardiac arrest requires the delivery of effective and high-quality chest compressions prior to the initiation of ECPR. The aim of this study was to evaluate and compare the effectiveness of mechanical and manual chest compressions until the initiation of ECPR. METHODS: This study was a secondary analysis of the Japanese retrospective multicenter registry "Study of Advanced Life Support for Ventricular Fibrillation by Extracorporeal Circulation II (SAVE-J II)". Patients were divided into two groups, one receiving mechanical chest compressions and the other receiving manual chest compressions. The primary outcome measure was mortality at hospital discharge, while the secondary outcome was the cerebral performance category (CPC) score at discharge. RESULTS: Of the 2157 patients enrolled in the SAVE-J II trial, 453 patients (329 in the manual compression group and 124 in the mechanical compression group) were included in the final analysis. Univariate analysis showed a significantly higher mortality rate at hospital discharge in the mechanical compression group compared to the manual compression group (odds ratio [95% CI] = 2.32 [1.34-4.02], p = 0.0026). Multivariate analysis showed that mechanical chest compressions were an independent factor associated with increased mortality at hospital discharge (adjusted odds ratio [95% CI] = 2.00 [1.11-3.58], p = 0.02). There was no statistically significant difference in CPC between the two groups. CONCLUSION: For patients with out-of-hospital cardiopulmonary arrest who require ECPR, extreme caution should be used when performing mechanical chest compressions.


Subject(s)
Cardiopulmonary Resuscitation , Out-of-Hospital Cardiac Arrest , Humans , Heart Massage , Out-of-Hospital Cardiac Arrest/therapy , Patient Discharge , Ventricular Fibrillation/therapy , Retrospective Studies
10.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34433666

ABSTRACT

Increasing attention has been paid to roles of tripartite motif-containing (TRIM) family proteins in cancer biology, often functioning as E3 ubiquitin ligases. In the present study, we focus on a contribution of TRIM47 to breast cancer biology, particularly to endocrine therapy resistance, which is a major clinical problem in breast cancer treatment. We performed immunohistochemical analysis of TRIM47 protein expression in 116 clinical samples of breast cancer patients with postoperative endocrine therapy using tamoxifen. Our clinicopathological study showed that higher immunoreactivity scores of TRIM47 were significantly associated with higher relapse rate of breast cancer patients (P = 0.012). As functional analyses, we manipulated TRIM47 expression in estrogen receptor-positive breast cancer cells MCF-7 and its 4-hydroxytamoxifen (OHT)-resistant derivative OHTR, which was established in a long-term culture with OHT. TRIM47 promoted both MCF-7 and OHTR cell proliferation. MCF-7 cells acquired tamoxifen resistance by overexpressing exogenous TRIM47. We found that TRIM47 enhances nuclear factor kappa-B (NF-κB) signaling, which further up-regulates TRIM47. We showed that protein kinase C epsilon (PKC-ε) and protein kinase D3 (PKD3), known as NF-κB-activating protein kinases, are directly associated with TRIM47 and stabilized in the presence of TRIM47. As an underlying mechanism, we showed TRIM47-dependent lysine 27-linked polyubiquitination of PKC-ε. These results indicate that TRIM47 facilitates breast cancer proliferation and endocrine therapy resistance by forming a ternary complex with PKC-ε and PKD3. TRIM47 and its associated kinases can be a potential diagnostic and therapeutic target for breast cancer refractory to endocrine therapy.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Drug Resistance, Neoplasm , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Tamoxifen/therapeutic use , Carrier Proteins/genetics , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , MCF-7 Cells , Multiprotein Complexes/metabolism , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Protein Kinase C/metabolism , Protein Kinase C-epsilon/metabolism , Protein Stability , Ubiquitination
11.
Int J Urol ; 31(6): 590-597, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38345202

ABSTRACT

Androgen deprivation therapy has achieved significant success in treating prostate cancer through strategies centered on the androgen receptor. However, the emergence of castration-resistant prostate cancer highlights this therapy limitation, underscoring the need to elucidate the mechanisms of treatment resistance. This review aimed to focus on multifaceted resistance mechanisms, including androgen receptor overexpression, splice variants, missense mutations, the involvement of the glucocorticoid receptor, and alterations in coregulators and transcription factors, revealing their roles in castration-resistant prostate cancer progression. These mechanisms promote cell survival and proliferation, depending on the androgen receptor signaling pathway, leading to resistance to conventional therapies. Amplification and mutations in the androgen receptor gene facilitate selective adaptation in treatment-resistant cells, consequently diminishing therapeutic efficacy. Furthermore, the activation of glucocorticoid receptors and aberrant regulation of specific coregulators and transcription factors contribute to the activation of androgen receptor-independent signaling pathways, promoting cell survival and proliferation. These findings hold promise for identifying new targets for treating castration-resistant prostate cancer and developing personalized treatment strategies. The development of future therapies will hinge on precisely targeting the androgen receptor signaling pathway, necessitating a deeper understanding of the molecular targets unique to castration-resistant prostate cancer.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Signal Transduction , Humans , Male , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Drug Resistance, Neoplasm/genetics , Cell Proliferation , Androgen Antagonists/therapeutic use , Gene Expression Regulation, Neoplastic , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Prostatic Neoplasms/therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics
12.
Int J Urol ; 31(1): 7-16, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37728330

ABSTRACT

Intraductal carcinoma of the prostate, a unique histopathologic entity that is often observed (especially in advanced prostate cancer), is characterized by the proliferation of malignant cells within normal acini or ducts surrounded by a basement membrane. Intraductal carcinoma of the prostate is almost invariably associated with an adjacent high-grade carcinoma and is occasionally observed as an isolated subtype. Intraductal carcinoma of the prostate has been demonstrated to be an independent poor prognostic factor for all stages of cancer, whether localized, de novo metastatic, or castration-resistant. It also has a characteristic genetic profile, including high genomic instability. Recognizing and differentiating it from other pathologies is therefore important in patient management, and morphological diagnostic criteria for intraductal carcinoma of the prostate have been established. This review summarizes and outlines the clinical and pathological features, differential diagnosis, molecular aspects, and management of intraductal carcinoma of the prostate, as described in previous studies. We also present a discussion and future perspectives regarding intraductal carcinoma of the prostate.


Subject(s)
Carcinoma, Intraductal, Noninfiltrating , Prostatic Neoplasms , Male , Humans , Prostate/pathology , Carcinoma, Intraductal, Noninfiltrating/diagnosis , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Diagnosis, Differential , Pelvis/pathology , Neoplasm Grading
13.
Gastroenterology ; 162(3): 799-812, 2022 03.
Article in English | MEDLINE | ID: mdl-34687740

ABSTRACT

BACKGROUND & AIMS: A detailed understanding of antitumor immunity is essential for optimal cancer immune therapy. Although defective mutations in the B2M and HLA-ABC genes, which encode molecules essential for antigen presentation, have been reported in several studies, the effects of these defects on tumor immunity have not been quantitatively evaluated. METHODS: Mutations in HLA-ABC genes were analyzed in 114 microsatellite instability-high colorectal cancers using a long-read sequencer. The data were further analyzed in combination with whole-exome sequencing, transcriptome sequencing, DNA methylation array, and immunohistochemistry data. RESULTS: We detected 101 truncating mutations in 57 tumors (50%) and loss of 61 alleles in 21 tumors (18%). Based on the integrated analysis that enabled the immunologic subclassification of microsatellite instability-high colorectal cancers, we identified a subtype of tumors in which lymphocyte infiltration was reduced, partly due to reduced expression of HLA-ABC genes in the absence of apparent genetic alterations. Survival time of patients with such tumors was shorter than in patients with other tumor types. Paradoxically, tumor mutation burden was highest in the subtype, suggesting that the immunogenic effect of accumulating mutations was counterbalanced by mutations that weakened immunoreactivity. Various genetic and epigenetic alterations, including frameshift mutations in RFX5 and promoter methylation of PSMB8 and HLA-A, converged on reduced expression of HLA-ABC genes. CONCLUSIONS: Our detailed immunogenomic analysis provides information that will facilitate the improvement and development of cancer immunotherapy.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Genes, MHC Class I/genetics , Tumor Escape/genetics , Tumor Escape/immunology , beta 2-Microglobulin/genetics , Alleles , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Methylation , Epigenesis, Genetic , Gene Expression , HLA-A Antigens/genetics , HLA-A Antigens/metabolism , Humans , Immunogenetics , Lymphocytes, Tumor-Infiltrating , Microsatellite Instability , Proteasome Endopeptidase Complex/genetics , Regulatory Factor X Transcription Factors/genetics , Survival Rate , beta 2-Microglobulin/metabolism
14.
J Bone Miner Metab ; 41(6): 817-821, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37525012

ABSTRACT

INTRODUCTION: Vitamin K is a fat-soluble vitamin discovered as an essential factor for blood coagulation. It is suggested that vitamin K can benefit several aging-related diseases, including osteoporosis, osteoarthritis, and dementia. We previously reported the cross-sectional association of vitamin K insufficiency with frailty in community-dwelling older adults. MATERIALS AND METHODS: In October 2020, a health examination of community-dwelling older adults (The Otassha Study) was performed, including frailty evaluation and blood tests. We used a ucOC and OC ratio (ucOC/OC) to indicate vitamin K insufficiency. One year later, we conducted a follow-up evaluation of frailty on 518 people who were not frail at baseline. The serum ucOC/OC at the baseline examination was divided into quartiles (Q1, Q2, Q3, and Q4). Odds ratio (OR) and 95% confidence interval (CI) were calculated using multivariate binary logistic regression for each quartile of ucOC/OC to determine the risk of incident frailty in the follow-up study, with the lowest quartile (Q1) as the reference. RESULTS: Among the 518 older adults who were not frail at baseline, 66 people (12.7%) became frail in the follow-up study. In the multivariate binary logistic regression analysis, setting the lowest quartile of ucOC/OC (Q1) as a reference, the OR of the incident frailty in the highest quartile (Q4) was 2.53 (95% CI 1.07, 4.92) which was significantly different from Q1. CONCLUSION: The findings of this longitudinal study suggest that vitamin K insufficiency has nutritional importance in predicting the future incidence of frailty in the Japanese older adult population.


Subject(s)
Frailty , Vitamin K , Humans , Aged , Longitudinal Studies , Incidence , Follow-Up Studies , Frailty/epidemiology , Independent Living , Cross-Sectional Studies , Osteocalcin
15.
Int J Urol ; 30(5): 464-471, 2023 05.
Article in English | MEDLINE | ID: mdl-36746652

ABSTRACT

OBJECTIVES: The coronavirus disease 2019 pandemic has affected cancer management worldwide. For upper tract urothelial carcinomas, delays in treatments are not recommended even during the pandemic. We investigated the impact of the pandemic on patients with these carcinomas who underwent radical nephroureterectomy (RNU) and adjuvant systematic therapy before and after COVID-19 spread in Japan. METHODS: This multicenter retrospective study included 304 patients who underwent RNU for upper tract urothelial carcinomas between May 1, 2019, and December 31, 2021, in Aichi, Japan. The patients were categorized into three groups based on whether they underwent surgery in the prepandemic (before infection spread in Japan), early pandemic (between confirmation of the first case and vaccination initiation), and late pandemic (after the start of vaccination in Japan) phases. The patient characteristics, diagnostic methods, pathological findings, and postoperative therapy were compared among the three phases. RESULTS: Overall, 74, 152, and 78 patients underwent RNU in the prepandemic, early pandemic, and late pandemic phases, respectively. The number of patients who underwent preoperative ureteroscopy decreased significantly from the prepandemic phase to the late pandemic phase due to pandemic-related restrictions (p = 0.016). There was no difference in the time to the first visit or pathological findings. Among patients classified as high-risk according to existing clinical trials, the proportion receiving adjuvant systematic therapy after RNU decreased significantly from 52.3% to 19% (p = 0.003). CONCLUSIONS: There was no difference in the pathological findings. The number of patients receiving appropriate adjuvant systematic therapy decreased during the pandemic.


Subject(s)
COVID-19 , Carcinoma, Transitional Cell , Ureteral Neoplasms , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/surgery , Nephroureterectomy/methods , Carcinoma, Transitional Cell/epidemiology , Carcinoma, Transitional Cell/surgery , Retrospective Studies , Japan/epidemiology , Pandemics/prevention & control , COVID-19/epidemiology , COVID-19/prevention & control , Ureteral Neoplasms/epidemiology , Ureteral Neoplasms/surgery , Ureteral Neoplasms/diagnosis
16.
Int J Mol Sci ; 24(3)2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36768177

ABSTRACT

Estrogen is an essential sex steroid hormone that functions primarily in female reproductive system, as well as in a variety of tissues and organs with pleiotropic effects, such as in cardiovascular, nervous, immune, and musculoskeletal systems. Women with low estrogen, as exemplified by those in postmenopause, are therefore prone to suffer from various disorders, i.e., cardiovascular disease, dementia, metabolic syndrome, osteoporosis, sarcopenia, frailty, and so on. Estrogen regulates the expression of its target genes by binding to its cognate receptors, estrogen receptors (ERs) α and ß. Notably, the estrogen-related receptors (ERRs) α, ß, and γ are originally identified as orphan receptors that share substantial structural homology and common transcriptional targets with ERs. Accumulating evidence suggests that ERs and ERRs play crucial roles in skeletal muscles, such as muscle mass maintenance, muscle exercise physiology, and muscle regeneration. In this article, we review potential regulatory roles of ERs and ERRs in muscle physiology, particularly with regard to mitochondrial function and metabolism.


Subject(s)
Muscular Diseases , Receptors, Estrogen , Female , Humans , Receptors, Estrogen/metabolism , Estrogens/metabolism , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Mitochondria/metabolism
17.
Int J Mol Sci ; 24(11)2023 May 28.
Article in English | MEDLINE | ID: mdl-37298366

ABSTRACT

The mitochondrial electron transport chain (ETC) plays an essential role in energy production by inducing oxidative phosphorylation (OXPHOS) to drive numerous biochemical processes in eukaryotic cells. Disorders of ETC and OXPHOS systems are associated with mitochondria- and metabolism-related diseases, including cancers; thus, a comprehensive understanding of the regulatory mechanisms of ETC and OXPHOS systems is required. Recent studies have indicated that noncoding RNAs (ncRNAs) play key roles in mitochondrial functions; in particular, some ncRNAs have been shown to modulate ETC and OXPHOS systems. In this review, we introduce the emerging roles of ncRNAs, including microRNAs (miRNAs), transfer-RNA-derived fragments (tRFs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), in the mitochondrial ETC and OXPHOS regulation.


Subject(s)
MicroRNAs , Neoplasms , RNA, Long Noncoding , Humans , Oxidative Phosphorylation , Electron Transport/genetics , RNA, Untranslated/genetics , MicroRNAs/genetics
18.
Biochem Biophys Res Commun ; 624: 81-88, 2022 10 08.
Article in English | MEDLINE | ID: mdl-35940131

ABSTRACT

Triple-negative breast cancer (TNBC) is characterized by its high ability of invasiveness and metastasis, namely lacking expression of estrogen receptor (ER), progesterone receptor, and HER2. We previously demonstrated that estrogen responsive finger protein (Efp) plays a tumor-promotive role in ER-positive breast cancer, yet it remains to be addressed whether Efp contributes to TNBC pathophysiology. We here found that Efp mRNA and protein were abundantly expressed in TNBC patient-derived cells and MDA-MB-231 cells. Efp silencing significantly decreased the growth and migration of both TNBC cell models. Cell-cycle profiling showed a decrease in the S phase population upon Efp silencing. Moreover, exogenous Efp expression increased the growth, migration, and the percentages of S phase population of TNBC cells. Transcriptomic analysis in the Efp-silenced TNBC cells identified several candidate Efp targets including cell cycle-related genes CDCA7 and HELLS, whose contribution to cell growth were validated by siRNA-mediated gene silencing. These results suggest that Efp plays a tumor-promotive role in TNBC and can be a potential therapeutic target for the aggressive disease.


Subject(s)
Triple Negative Breast Neoplasms , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Profiling , Humans , Nuclear Proteins/genetics , Transcription Factors , Tripartite Motif Proteins , Triple Negative Breast Neoplasms/pathology , Ubiquitin-Protein Ligases
19.
Biochem Biophys Res Commun ; 628: 11-17, 2022 11 05.
Article in English | MEDLINE | ID: mdl-36063597

ABSTRACT

Estrogen is a female hormone that plays a role in various tissues, although the mechanism in skeletal muscle has not been fully clarified. We previously showed that systemic administration of estrogen for 10 weeks ameliorated decreased exercise endurance in ovariectomized mice. To assess whether a long-term and muscle-specific activation of estrogen signaling modulates muscle function, we constructed an expression plasmid for a constitutively active estrogen receptor α (caERα) under the control of muscle creatine kinase (Mck) gene promoter/enhancer. In C2C12 mouse myoblastic cells, transfection of the Mck-caERα plasmid elevated the estrogen response element-driven transcription in a ligand-independent manner. Using this construct, we generated Mck-caERα transgenic mice, in which caERα is predominantly expressed in muscle. Treadmill running test revealed that female Mck-caERα mice exhibit a prolonged running time and distance compared with the wild-type mice. Moreover, microarray expression analysis revealed that the genes related to lipid metabolism, insulin signaling, and growth factor signaling were particularly upregulated in the quadriceps femoris muscle of Mck-caERα mice. These results suggest that estrogen signaling potentiates exercise endurance in skeletal muscle through modulating the expression of metabolism-associated genes.


Subject(s)
Estrogen Receptor alpha , Physical Endurance , Animals , Creatine Kinase, MM Form/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Insulins/metabolism , Ligands , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Physical Endurance/genetics
20.
Biochem Biophys Res Commun ; 623: 9-16, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35868070

ABSTRACT

Androgens and androgen receptor (AR) have a central role in prostate cancer progression by regulating its downstream signaling. Although androgen depletion therapy (ADT) is the primary treatment for most prostate cancers, they acquires resistance to ADT and become castration resistant prostate cancers (CRPC). AR complex formation with multiple transcription factors is important for enhancer activity and transcriptional regulation, which can contribute to cancer progression and resistance to ADT. We previously demonstrated that OCT1 collaborates with AR in prostate cancer, and that a pyrrole-imidazole (PI) polyamide (PIP) targeting OCT1 inhibits cell and castration-resistant tumor growth (Obinata D et al. Oncogene 2016). PIP can bind to DNA non-covalently without a drug delivery system unlike most DNA targeted therapeutics. In the present study, we developed a PIP modified with a DNA alkylating agent, chlorambucil (ChB) (OCT1-PIP-ChB). Then its effect on the growth of prostate cancer LNCaP, 22Rv1, and PC3 cells, pancreatic cancer BxPC3 cells, and colon cancer HCT116 cells, as well as non-cancerous MCF-10A epithelial cells, were analyzed. It was shown that the IC50s of OCT1-PIP-ChB for 22Rv1 and LNCaP were markedly lower compared to other cells, including non-cancerous MCF-10A cells. Comprehensive gene expression analysis of CRPC model 22Rv1 cells treated with IC50 concentrations of OCT1-PIP-ChB revealed that the gene group involved in DNA double-strand break repair was the most enriched among gene sets repressed by OCT1-PIP-ChB treatment. Importantly, in vivo study using 22Rv1 xenografts, we showed that OCT1-PIP-ChB significantly reduced tumor growth compared to the control group without showing obvious adverse effects. Thus, the PIP combined with ChB can exert a significant inhibitory effect on prostate cancer cell proliferation and castration-resistant tumor growth, suggesting a potential role as a therapeutic agent.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Alkylating Agents , Cell Line, Tumor , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Male , Nylons/pharmacology , Prostatic Neoplasms, Castration-Resistant/pathology , Pyrroles/pharmacology , Pyrroles/therapeutic use , Receptors, Androgen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL