99851
|
DHULI KRISTJANA, BONETTI GABRIELE, ANPILOGOV KYRYLO, HERBST KARENL, CONNELLY STEPHENTHADDEUS, BELLINATO FRANCESCO, GISONDI PAOLO, BERTELLI MATTEO. Validating methods for testing natural molecules on molecular pathways of interest in silico and in vitro. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2022; 63:E279-E288. [PMID: 36479497 PMCID: PMC9710400 DOI: 10.15167/2421-4248/jpmh2022.63.2s3.2770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Differentially expressed genes can serve as drug targets and are used to predict drug response and disease progression. In silico drug analysis based on the expression of these genetic biomarkers allows the detection of putative therapeutic agents, which could be used to reverse a pathological gene expression signature. Indeed, a set of bioinformatics tools can increase the accuracy of drug discovery, helping in biomarker identification. Once a drug target is identified, in vitro cell line models of disease are used to evaluate and validate the therapeutic potential of putative drugs and novel natural molecules. This study describes the development of efficacious PCR primers that can be used to identify gene expression of specific genetic pathways, which can lead to the identification of natural molecules as therapeutic agents in specific molecular pathways. For this study, genes involved in health conditions and processes were considered. In particular, the expression of genes involved in obesity, xenobiotics metabolism, endocannabinoid pathway, leukotriene B4 metabolism and signaling, inflammation, endocytosis, hypoxia, lifespan, and neurotrophins were evaluated. Exploiting the expression of specific genes in different cell lines can be useful in in vitro to evaluate the therapeutic effects of small natural molecules.
Collapse
Affiliation(s)
- KRISTJANA DHULI
- MAGI’S LAB, Rovereto (TN), Italy
- Correspondence: Kristjana Dhuli, MAGI’S LAB, Rovereto (TN), 38068, Italy. E-mail:
| | | | | | - KAREN L. HERBST
- Total Lipedema Care, Beverly Hills California and Tucson Arizona, USA
| | - STEPHEN THADDEUS CONNELLY
- San Francisco Veterans Affairs Health Care System, Department of Oral & Maxillofacial Surgery, University of California, San Francisco, CA, USA7
| | - FRANCESCO BELLINATO
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - PAOLO GISONDI
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - MATTEO BERTELLI
- MAGI’S LAB, Rovereto (TN), Italy
- MAGI EUREGIO, Bolzano, BZ, Italy
- MAGISNAT, Peachtree Corners (GA), USA
| |
Collapse
|
99852
|
Necula L, Matei L, Dragu D, Pitica I, Neagu A, Bleotu C, Diaconu CC, Chivu-Economescu M. Collagen Family as Promising Biomarkers and Therapeutic Targets in Cancer. Int J Mol Sci 2022; 23:12415. [PMID: 36293285 PMCID: PMC9604126 DOI: 10.3390/ijms232012415] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Despite advances in cancer detection and therapy, it has been estimated that the incidence of cancers will increase, while the mortality rate will continue to remain high, a fact explained by the large number of patients diagnosed in advanced stages when therapy is often useless. Therefore, it is necessary to invest knowledge and resources in the development of new non-invasive biomarkers for the early detection of cancer and new therapeutic targets for better health management. In this review, we provided an overview on the collagen family as promising biomarkers and on how they may be exploited as therapeutic targets in cancer. The collagen family tridimensional structure, organization, and functions are very complex, being in a tight relationship with the extracellular matrix, tumor, and immune microenvironment. Moreover, accumulating evidence underlines the role of collagens in promoting tumor growth and creating a permissive tumor microenvironment for metastatic dissemination. Knowledge of the molecular basis of these interactions may help in cancer diagnosis and prognosis, in overcoming chemoresistance, and in providing new targets for cancer therapies.
Collapse
Affiliation(s)
- Laura Necula
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania
- Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania
| | - Lilia Matei
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania
| | - Denisa Dragu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania
| | - Ioana Pitica
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania
| | - Ana Neagu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania
| | - Carmen C. Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania
| | - Mihaela Chivu-Economescu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania
| |
Collapse
|
99853
|
Lang R, Li H, Luo X, Liu C, Zhang Y, Guo S, Xu J, Bao C, Dong W, Yu Y. Expression and mechanisms of interferon-stimulated genes in viral infection of the central nervous system (CNS) and neurological diseases. Front Immunol 2022; 13:1008072. [PMID: 36325336 PMCID: PMC9618809 DOI: 10.3389/fimmu.2022.1008072] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/28/2022] [Indexed: 09/16/2023] Open
Abstract
Interferons (IFNs) bind to cell surface receptors and activate the expression of interferon-stimulated genes (ISGs) through intracellular signaling cascades. ISGs and their expression products have various biological functions, such as antiviral and immunomodulatory effects, and are essential effector molecules for IFN function. ISGs limit the invasion and replication of the virus in a cell-specific and region-specific manner in the central nervous system (CNS). In addition to participating in natural immunity against viral infections, studies have shown that ISGs are essential in the pathogenesis of CNS disorders such as neuroinflammation and neurodegenerative diseases. The aim of this review is to present a macroscopic overview of the characteristics of ISGs that restrict viral neural invasion and the expression of the ISGs underlying viral infection of CNS cells. Furthermore, we elucidate the characteristics of ISGs expression in neurological inflammation, neuropsychiatric disorders such as depression as well as neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Finally, we summarize several ISGs (ISG15, IFIT2, IFITM3) that have been studied more in recent years for their antiviral infection in the CNS and their research progress in neurological diseases.
Collapse
Affiliation(s)
- Rui Lang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huiting Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xiaoqin Luo
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Cencen Liu
- Department of Pathology, People’s Hospital of Zhongjiang County, DeYang, China
| | - Yiwen Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - ShunYu Guo
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jingyi Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Changshun Bao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Neurological diseases and brain function laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yang Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
99854
|
Altintas DM, Gallo S, Basilico C, Cerqua M, Bocedi A, Vitacolonna A, Botti O, Casanova E, Rancati I, Milanese C, Notari S, Gambardella G, Ricci G, Mastroberardino PG, Boccaccio C, Crepaldi T, Comoglio PM. The PSI Domain of the MET Oncogene Encodes a Functional Disulfide Isomerase Essential for the Maturation of the Receptor Precursor. Int J Mol Sci 2022; 23:ijms232012427. [PMID: 36293286 PMCID: PMC9604360 DOI: 10.3390/ijms232012427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 11/29/2022] Open
Abstract
The tyrosine kinase receptor encoded by the MET oncogene has been extensively studied. Surprisingly, one extracellular domain, PSI, evolutionary conserved between plexins, semaphorins, and integrins, has no established function. The MET PSI sequence contains two CXXC motifs, usually found in protein disulfide isomerases (PDI). Using a scrambled oxidized RNAse enzymatic activity assay in vitro, we show, for the first time, that the MET extracellular domain displays disulfide isomerase activity, abolished by PSI domain antibodies. PSI domain deletion or mutations of CXXC sites to AXXA or SXXS result in a significant impairment of the cleavage of the MET 175 kDa precursor protein, abolishing the maturation of α and β chains, of, respectively, 50 kDa and 145 kDa, disulfide-linked. The uncleaved precursor is stuck in the Golgi apparatus and, interestingly, is constitutively phosphorylated. However, no signal transduction is observed as measured by AKT and MAPK phosphorylation. Consequently, biological responses to the MET ligand—hepatocyte growth factor (HGF)—such as growth and epithelial to mesenchymal transition, are hampered. These data show that the MET PSI domain is functional and is required for the maturation, surface expression, and biological functions of the MET oncogenic protein.
Collapse
Affiliation(s)
- Dogus Murat Altintas
- IFOM, FIRC Institute for Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
- Correspondence: (D.M.A.); (P.M.C.)
| | - Simona Gallo
- Department of Oncology, University of Turin, 10060 Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, Italy
| | | | - Marina Cerqua
- IFOM, FIRC Institute for Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
| | - Alessio Bocedi
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Annapia Vitacolonna
- Department of Oncology, University of Turin, 10060 Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, Italy
| | - Orsola Botti
- IFOM, FIRC Institute for Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
| | - Elena Casanova
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, Italy
| | - Ilaria Rancati
- IFOM, FIRC Institute for Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
| | - Chiara Milanese
- IFOM, FIRC Institute for Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
| | - Sara Notari
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Giorgia Gambardella
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Giorgio Ricci
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Pier Giorgio Mastroberardino
- IFOM, FIRC Institute for Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
- Department of Life, Health, and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Carla Boccaccio
- Department of Oncology, University of Turin, 10060 Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, Italy
| | - Tiziana Crepaldi
- Department of Oncology, University of Turin, 10060 Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, Italy
| | - Paolo Maria Comoglio
- IFOM, FIRC Institute for Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
- Correspondence: (D.M.A.); (P.M.C.)
| |
Collapse
|
99855
|
Tabata J, Nakaoku T, Araki M, Yoshino R, Kohsaka S, Otsuka A, Ikegami M, Ui A, Kanno SI, Miyoshi K, Matsumoto S, Sagae Y, Yasui A, Sekijima M, Mano H, Okuno Y, Okamoto A, Kohno T. Novel Calcium-Binding Ablating Mutations Induce Constitutive RET Activity and Drive Tumorigenesis. Cancer Res 2022; 82:3751-3762. [PMID: 36166639 PMCID: PMC9574375 DOI: 10.1158/0008-5472.can-22-0834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 07/13/2022] [Accepted: 08/11/2022] [Indexed: 01/07/2023]
Abstract
Distinguishing oncogenic mutations from variants of unknown significance (VUS) is critical for precision cancer medicine. Here, computational modeling of 71,756 RET variants for positive selection together with functional assays of 110 representative variants identified a three-dimensional cluster of VUSs carried by multiple human cancers that cause amino acid substitutions in the calmodulin-like motif (CaLM) of RET. Molecular dynamics simulations indicated that CaLM mutations decrease interactions between Ca2+ and its surrounding residues and induce conformational distortion of the RET cysteine-rich domain containing the CaLM. RET-CaLM mutations caused ligand-independent constitutive activation of RET kinase by homodimerization mediated by illegitimate disulfide bond formation. RET-CaLM mutants possessed oncogenic and tumorigenic activities that could be suppressed by tyrosine kinase inhibitors targeting RET. This study identifies calcium-binding ablating mutations as a novel type of oncogenic mutation of RET and indicates that in silico-driven annotation of VUSs of druggable oncogenes is a promising strategy to identify targetable driver mutations. SIGNIFICANCE Comprehensive proteogenomic and in silico analyses of a vast number of VUSs identify a novel set of oncogenic and druggable mutations in the well-characterized RET oncogene.
Collapse
Affiliation(s)
- Junya Tabata
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan.,Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Nakaoku
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan.,Corresponding Authors: Takashi Nakaoku, Division of Genome Biology, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan. Phone: 813-3542-2511; E-mail: ; and Takashi Kohno, Division of Genome Biology, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan. Phone: 813-3547-5272; E-mail:
| | - Mitsugu Araki
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryunosuke Yoshino
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Ayaka Otsuka
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Masachika Ikegami
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Ayako Ui
- Department of Molecular Oncology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Shin-ichiro Kanno
- Department of Molecular Oncology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Keiko Miyoshi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | | | - Yukari Sagae
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Yasui
- IDAC Fellow Laboratory, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Masakazu Sekijima
- Department of Computer Science, Tokyo Institute of Technology, Yokohama, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasushi Okuno
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan.,Corresponding Authors: Takashi Nakaoku, Division of Genome Biology, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan. Phone: 813-3542-2511; E-mail: ; and Takashi Kohno, Division of Genome Biology, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan. Phone: 813-3547-5272; E-mail:
| |
Collapse
|
99856
|
Cheng X, Dai C, Wen Y, Wang X, Bo X, He S, Peng S. NeRD: a multichannel neural network to predict cellular response of drugs by integrating multidimensional data. BMC Med 2022; 20:368. [PMID: 36244991 PMCID: PMC9575288 DOI: 10.1186/s12916-022-02549-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Considering the heterogeneity of tumors, it is a key issue in precision medicine to predict the drug response of each individual. The accumulation of various types of drug informatics and multi-omics data facilitates the development of efficient models for drug response prediction. However, the selection of high-quality data sources and the design of suitable methods remain a challenge. METHODS In this paper, we design NeRD, a multidimensional data integration model based on the PRISM drug response database, to predict the cellular response of drugs. Four feature extractors, including drug structure extractor (DSE), molecular fingerprint extractor (MFE), miRNA expression extractor (mEE), and copy number extractor (CNE), are designed for different types and dimensions of data. A fully connected network is used to fuse all features and make predictions. RESULTS Experimental results demonstrate the effective integration of the global and local structural features of drugs, as well as the features of cell lines from different omics data. For all metrics tested on the PRISM database, NeRD surpassed previous approaches. We also verified that NeRD has strong reliability in the prediction results of new samples. Moreover, unlike other algorithms, when the amount of training data was reduced, NeRD maintained stable performance. CONCLUSIONS NeRD's feature fusion provides a new idea for drug response prediction, which is of great significance for precise cancer treatment.
Collapse
Affiliation(s)
- Xiaoxiao Cheng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Chong Dai
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.,Department of Biotechnology, Beijing Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Yuqi Wen
- Department of Biotechnology, Beijing Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Xiaoqi Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Xiaochen Bo
- Department of Biotechnology, Beijing Institute of Health Service and Transfusion Medicine, Beijing, China.
| | - Song He
- Department of Biotechnology, Beijing Institute of Health Service and Transfusion Medicine, Beijing, China.
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China. .,The State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China.
| |
Collapse
|
99857
|
Habibi M, Taheri G. A new machine learning method for cancer mutation analysis. PLoS Comput Biol 2022; 18:e1010332. [PMID: 36251702 PMCID: PMC9612828 DOI: 10.1371/journal.pcbi.1010332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/27/2022] [Accepted: 10/05/2022] [Indexed: 11/23/2022] Open
Abstract
It is complicated to identify cancer-causing mutations. The recurrence of a mutation in patients remains one of the most reliable features of mutation driver status. However, some mutations are more likely to happen than others for various reasons. Different sequencing analysis has revealed that cancer driver genes operate across complex pathways and networks, with mutations often arising in a mutually exclusive pattern. Genes with low-frequency mutations are understudied as cancer-related genes, especially in the context of networks. Here we propose a machine learning method to study the functionality of mutually exclusive genes in the networks derived from mutation associations, gene-gene interactions, and graph clustering. These networks have indicated critical biological components in the essential pathways, especially those mutated at low frequency. Studying the network and not just the impact of a single gene significantly increases the statistical power of clinical analysis. The proposed method identified important driver genes with different frequencies. We studied the function and the associated pathways in which the candidate driver genes participate. By introducing lower-frequency genes, we recognized less studied cancer-related pathways. We also proposed a novel clustering method to specify driver modules. We evaluated each driver module with different criteria, including the terms of biological processes and the number of simultaneous mutations in each cancer. Materials and implementations are available at: https://github.com/MahnazHabibi/MutationAnalysis.
Collapse
Affiliation(s)
- Mahnaz Habibi
- Department of Mathematics, Qazvin Branch, Islamic Azad University, Qazvin, Iran
| | - Golnaz Taheri
- Department of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden
- Science for Life Laboratory, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
99858
|
Xiong Z, Wu S, Li FJ, Luo C, Jin QY, Connolly ID, Hayden Gephart M, You L. Elevated ETV6 Expression in Glioma Promotes an Aggressive In Vitro Phenotype Associated with Shorter Patient Survival. Genes (Basel) 2022; 13:genes13101882. [PMID: 36292767 PMCID: PMC9656946 DOI: 10.3390/genes13101882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/29/2022] Open
Abstract
Background: GBM astrocytes may adopt fetal astrocyte transcriptomic signatures involved in brain development and migration programs to facilitate diffuse tumor infiltration. Our previous data show that ETS variant 6 (ETV6) is highly expressed in human GBM and fetal astrocytes compared to normal mature astrocytes. We hypothesized that ETV6 played a role in GBM tumor progression. Methods: Expression of ETV6 was first examined in two American and three Chinese tissue microarrays. The correlation between ETV6 staining intensity and patient survival was calculated, followed by validation using public databases—TCGA and REMBRANDT. The effect of ETV6 knockdown on glioma cell proliferation (EdU), viability (AnnexinV labeling), clonogenic growth (colony formation), and migration/invasion (transwell assays) in GBM cells was tested. RNA sequencing and Western blot were performed to elucidate the underlying molecular mechanisms. Results: ETV6 was highly expressed in GBM and associated with an unfavorable prognosis. ETV6 silencing in glioma cells led to increased apoptosis or decreased proliferation, clonogenicity, migration, and invasion. RNA-Seq-based gene expression and pathway analyses revealed that ETV6 knockdown in U251 cells led to the upregulation of genes involved in extracellular matrix organization, NF-κB signaling, TNF-mediated signaling, and the downregulation of genes in the regulation of cell motility, cell proliferation, PI3K-AKT signaling, and the Ras pathway. The downregulation of the PI3K-AKT and Ras-MAPK pathways were further validated by immunoblotting. Conclusion: Our findings suggested that ETV6 was highly expressed in GBM and its high expression correlated with poor survival. ETV6 silencing decreased an aggressive in vitro phenotype probably via the PI3K-AKT and Ras-MAPK pathways. The study encourages further investigation of ETV6 as a potential therapeutic target of GBM.
Collapse
Affiliation(s)
- Zhang Xiong
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
- Neurosurgical Institute, Fudan University, Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai 200040, China
| | - Shuai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
- Neurosurgical Institute, Fudan University, Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai 200040, China
| | - Feng-jiao Li
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Chen Luo
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
- Neurosurgical Institute, Fudan University, Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai 200040, China
| | - Qiu-yan Jin
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ian David Connolly
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Melanie Hayden Gephart
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
- Correspondence: to: (M.H.G.); (L.Y.)
| | - Linya You
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai 200032, China
- Correspondence: to: (M.H.G.); (L.Y.)
| |
Collapse
|
99859
|
Zhang C, Kang T, Wang X, Song J, Zhang J, Li G. Stimuli-responsive platinum and ruthenium complexes for lung cancer therapy. Front Pharmacol 2022; 13:1035217. [PMID: 36324675 PMCID: PMC9618881 DOI: 10.3389/fphar.2022.1035217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
Lung cancer is the most common cause of cancer-related deaths worldwide. More efficient treatments are desperately needed. For decades, the success of platinum-based anticancer drugs has promoted the exploration of metal-based agents. Four ruthenium-based complexes have also entered clinical trials as candidates of anticancer metallodrugs. However, systemic toxicity, severe side effects and drug-resistance impeded their applications and efficacy. Stimuli-responsiveness of Pt- and Ru-based complexes provide a great chance to weaken the side effects and strengthen the clinical efficacy in drug design. This review provides an overview on the stimuli-responsive Pt- and Ru-based metallic anticancer drugs for lung cancer. They are categorized as endo-stimuli-responsive, exo-stimuli-responsive, and dual-stimuli-responsive prodrugs based on the nature of stimuli. We describe various representative examples of structure, response mechanism, and potential medical applications in lung cancer. In the end, we discuss the future opportunities and challenges in this field.
Collapse
Affiliation(s)
- Cheng Zhang
- The Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Tong Kang
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xinyi Wang
- The Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jiaqi Song
- Department of Biophysics, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jia Zhang
- The Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Jia Zhang, ; Guanying Li,
| | - Guanying Li
- Department of Biophysics, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Jia Zhang, ; Guanying Li,
| |
Collapse
|
99860
|
Newest Therapies for Cholangiocarcinoma: An Updated Overview of Approved Treatments with Transplant Oncology Vision. Cancers (Basel) 2022; 14:cancers14205074. [PMID: 36291857 PMCID: PMC9600404 DOI: 10.3390/cancers14205074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Cholangiocarcinoma is a relatively rare but deadly disease with traditionally limited treatment options. The disease can be categorized by anatomic location within the biliary tree, with different associated risk factors and molecular profiles. Recent years have seen a burgeoning of targeted therapies that have enhanced survival in subsets of patients with certain mutations. We herein discuss these more recent advances as well as providing an overview of more well-known treatment modalities, with the goal of providing an accessible source for practicing clinicians. Abstract A minority of cholangiocarcinoma (CCA) can be cured by surgical intervention (i.e., liver resection (LR) and liver transplantation (LT)). When modern criteria for LT are met, this intervention along with neoadjuvant treatments may achieve unprecedented survival in selected patients. Liver resection is associated with a median overall survival (OS) of 40 months, this number drastically decreases for unresectable advanced cholangiocarcinoma (CCA), which is treated with systemic therapy. The first-line chemotherapy regimen of gemcitabine and cisplatin is associated with a median overall survival of only 11.7 months. Since the Food and Drug Administration (FDA)’s approval of the isocitrate dehydrogenase (IDH) 1 inhibitor ivosidenib in August 2021, there has been increasing interest in targeted therapy for CCA patients harboring mutations in fibroblast growth factor receptor (FGFR) 2, neurotrophic receptor tyrosine kinase (NTRK), B-raf kinase (BRAF), and HER2. At the same time, immunotherapy with immune checkpoint inhibitors isalso being used in relapsed CCA. This review looks into the most recently completed and ongoing studies of targeted therapy as monotherapy or in combination with chemo- and/or immunotherapy. Whether it is resection, liver transplant, radiotherapy, chemotherapy, immunotherapy, targeted therapy, or any combination of these treatment modalities, great strides are being made to improve outcomes for this challenging disease.
Collapse
|
99861
|
Liu J, Wang H, Zhang M, Li Y, Wang R, Chen H, Wang B, Gao X, Song S, Wang Y, Ren Y, Li J, Liu P. Metformin and simvastatin synergistically suppress endothelin 1-induced hypoxia and angiogenesis in multiple cancer types. Cancer Sci 2022; 114:640-653. [PMID: 36156330 PMCID: PMC9899631 DOI: 10.1111/cas.15602] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/08/2022] [Accepted: 09/20/2022] [Indexed: 01/07/2023] Open
Abstract
Multiple cancers have been reported to be associated with angiogenesis and are sensitive to anti-angiogenic therapies. Vascular normalization, by restoring proper tumor perfusion and oxygenation, could limit tumor cell invasiveness and improve the effectiveness of anticancer treatments. However, the underlying anticancer mechanisms of antiangiogenic drugs are still unknown. Metformin (MET) and simvastatin (SVA), two metabolic-related drugs, have been shown to play important roles in modulating the hypoxic tumor microenvironment and angiogenesis. Whether the combination of MET and SVA could exert a more effective antitumor effect than individual treatments has not been examined. The antitumor effect of the synergism of SVA and MET was detected in mouse models, breast cancer patient-derived organoids, and multiple tumor cell lines compared with untreated, SVA, or MET alone. RNA sequencing revealed that the combination of MET and SVA (but not MET or SVA alone) inhibited the expression of endothelin 1 (ET-1), an important regulator of angiogenesis and the hypoxia-related pathway. We demonstrate that the MET and SVA combination showed synergistic effects on inhibiting tumor cell proliferation, promoting apoptosis, alleviating hypoxia, decreasing angiogenesis, and increasing vessel normalization compared with the use of a single agent alone. The MET and SVA combination suppressed ET-1-induced hypoxia-inducible factor 1α expression by increasing prolyl hydroxylase 2 (PHD2) expression. Furthermore, the MET and SVA combination showed a more potent anticancer effect compared with bosentan. Together, our findings suggest the potential application of the MET and SVA combination in antitumor therapy.
Collapse
Affiliation(s)
- Jie Liu
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Huxia Wang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Mammary DepartmentShaanxi Provincial Cancer HospitalXi'anShaanxi ProvinceChina
| | - Miao Zhang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Yazhao Li
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Ruiqi Wang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - He Chen
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Bo Wang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Xiaoqian Gao
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Shaoran Song
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Yaochun Wang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Yu Ren
- Department of Surgical OncologyThe First Affiliated Hospital of Xi' an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Juan Li
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Peijun Liu
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina,Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| |
Collapse
|
99862
|
Guerriaud M, Kohli E. RNA-based drugs and regulation: Toward a necessary evolution of the definitions issued from the European union legislation. Front Med (Lausanne) 2022; 9:1012497. [PMID: 36325384 PMCID: PMC9618588 DOI: 10.3389/fmed.2022.1012497] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/26/2022] [Indexed: 11/14/2022] Open
Abstract
Many RNA-based drugs, both vaccines and non-vaccines, are under development or even approved. They include coding mRNAs and non-coding (nc) RNAs among them antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), micro-RNAs (miRNAs), small activating RNAs (saRNAs), RNA aptamers and RNA guides. According to the European Union (EU) legislation, these products can be currently categorized into different regulatory statuses, depending, for vaccines, on their target (infectious disease or not) and, for other drugs, on how they are obtained (chemically or biologically). This classification is fundamental to the type of marketing authorization (MA), and therefore to the controls to be performed, from preclinical stages through clinical trials to pharmacovigilance, to meet the safety requirements for patients. However, the current rules raise several problems, in particular the risk, because technology is evolving, to have similar RNA drugs being covered by very different legal statuses and the lack of international harmonization. The objectives of this study are (i) to review how RNA medicinal products are currently legally categorized in the EU and especially whether they fall under the status of gene therapy medicinal products (GTMP), a regulatory status belonging to advanced therapy medicinal products (ATMP), (ii) to discuss the issues generated by this classification, with a focus on the heterogeneity of statuses of these products, the differences with the American and ICH definitions and the potential impact on the safety requirements.
Collapse
Affiliation(s)
- Mathieu Guerriaud
- CREDIMI Laboratory EA 7532 and Laboratory of Excellence LipSTIC ANR-11-LABX-0021, Faculty of Health Sciences (Pharmacy), University of Burgundy, Dijon, France
- *Correspondence: Mathieu Guerriaud,
| | - Evelyne Kohli
- UMR INSERM/uB/AGROSUP 1231, Team 3 HSP-Pathies, Labelled Ligue Nationale Contre le Cancer and Laboratory of Excellence LipSTIC ANR-11-LABX-0021, Faculty of Health Sciences (Pharmacy), University of Burgundy, Dijon, France
| |
Collapse
|
99863
|
Sophiya P, Urs D, K. Lone J, Giresha AS, Krishna Ram H, Manjunatha JG, El-Serehy HA, Narayanappa M, Shankar J, Bhardwaj R, Ahmad Guru S, Dharmappa KK. Quercitrin neutralizes sPLA2IIa activity, reduces the inflammatory IL-6 level in PC3 cell lines, and exhibits anti-tumor activity in the EAC-bearing mice model. Front Pharmacol 2022; 13:996285. [PMID: 36324674 PMCID: PMC9620381 DOI: 10.3389/fphar.2022.996285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/15/2022] [Indexed: 04/12/2024] Open
Abstract
Human phospholipase A2 group IIa (sPLA2IIa) is an inflammatory enzyme that plays a significant role in tumorigenesis. Inhibiting the sPLA2IIa enzyme with an effective molecule can reduce the inflammatory response and halt cancer progression. The present study evaluates quercitrin, a biflavonoid, for sPLA2IIa inhibition and anticancer activity. Quercitrin inhibited sPLA2IIa activity to a greater extent-at 86.24% ± 1.41 with an IC50 value of 8.77 μM ± 0.9. The nature of sPLA2IIa inhibition was evaluated by increasing calcium concentration from 2.5 to 15 µM and substrate from 20 to 120 nM, which did not alter the level of inhibition. Intrinsic fluorescence and far UV-CD studies confirmed the direct interaction of quercitrin with the sPLA2IIa enzyme. This significantly reduced the sPLA2IIa-induced hemolytic activity and mouse paw edema from 97.32% ± 1.23-16.91% ± 2.03 and 172.87% ± 1.9-118.41% ± 2.53, respectively. As an anticancer activity, quercitrin reduced PC-3 cell viability from 98.66% ± 2.51-18.3% ± 1.52 and significantly decreased the IL-6 level in a dose-dependent manner from 98.35% ± 2.2-37.12% ± 2.4. It increased the mean survival time (MST) of EAC-bearing Swiss albino mice from 30 to 35 days. It obeyed Lipinski's rule of five, suggesting a druggable property. Thus, all the above experimental results were promising and encouraged further investigation into developing quercitrin as a therapeutic drug for both inflammatory diseases and cancers.
Collapse
Affiliation(s)
- P. Sophiya
- Inflammation Research Laboratory, Department of Studies and Research in Biochemistry, Jnana Kaveri Post Graduate campus, Mangalore University, Kushalanagar, India
| | - Deepadarshan Urs
- Inflammation Research Laboratory, Department of Studies and Research in Biochemistry, Jnana Kaveri Post Graduate campus, Mangalore University, Kushalanagar, India
| | - Jafar K. Lone
- ICAR-National Bureau of Plant Genetic Resources, New Delhi, India
| | - A. S. Giresha
- Department of Biochemistry, School of Science, Jain (Deemed-to-be University), Bangalore, India
| | - H. Krishna Ram
- Nisarga Research and Development Trust (T), Bengaluru, India
| | - J. G. Manjunatha
- Department of Chemistry, FMKMC College, Mangalore University Constituent College, Madikeri, India
| | - Hamed A. El-Serehy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - M. Narayanappa
- Inflammation Research Laboratory, Department of Studies and Research in Biochemistry, Jnana Kaveri Post Graduate campus, Mangalore University, Kushalanagar, India
| | - J. Shankar
- Department of Studies in Food Technology, Davanagere University, Davanagere, India
| | - Ragini Bhardwaj
- Department of Microbiology and Biotechnology, Banasthali Vidyapith, Jaipur, India
| | - Sameer Ahmad Guru
- Department of Development of Biology and Regenerative Medicine, Lurie Children Hospital, Northwestern University, Chicago, IL, United States
| | - K. K. Dharmappa
- Inflammation Research Laboratory, Department of Studies and Research in Biochemistry, Jnana Kaveri Post Graduate campus, Mangalore University, Kushalanagar, India
| |
Collapse
|
99864
|
Feng S, Lou K, Luo C, Zou J, Zou X, Zhang G. Obesity-Related Cross-Talk between Prostate Cancer and Peripheral Fat: Potential Role of Exosomes. Cancers (Basel) 2022; 14:5077. [PMID: 36291860 PMCID: PMC9600017 DOI: 10.3390/cancers14205077] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/06/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022] Open
Abstract
The molecular mechanisms of obesity-induced cancer progression have been extensively explored because of the significant increase in obesity and obesity-related diseases worldwide. Studies have shown that obesity is associated with certain features of prostate cancer. In particular, bioactive factors released from periprostatic adipose tissues mediate the bidirectional communication between periprostatic adipose tissue and prostate cancer. Moreover, recent studies have shown that extracellular vesicles have a role in the relationship between tumor peripheral adipose tissue and cancer progression. Therefore, it is necessary to investigate the feedback mechanisms between prostate cancer and periglandular adipose and the role of exosomes as mediators of signal exchange to understand obesity as a risk factor for prostate cancer. This review summarizes the two-way communication between prostate cancer and periglandular adipose and discusses the potential role of exosomes as a cross-talk and the prospect of using adipose tissue as a means to obtain exosomes in vitro. Therefore, this review may provide new directions for the treatment of obesity to suppress prostate cancer.
Collapse
Affiliation(s)
- Shangzhi Feng
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Kecheng Lou
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Cong Luo
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Junrong Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Institute of Urology, The First Affiliated Hospital of Ganna Medical University, Ganzhou 341000, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou 341000, China
| | - Xiaofeng Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Institute of Urology, The First Affiliated Hospital of Ganna Medical University, Ganzhou 341000, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou 341000, China
| | - Guoxi Zhang
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Institute of Urology, The First Affiliated Hospital of Ganna Medical University, Ganzhou 341000, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou 341000, China
| |
Collapse
|
99865
|
Spectrum of EGFR mutation and its relation with high-risk predictors in thyroid cancer in Kashmiri population: 2 years prospective study at a tertiary care hospital. J Egypt Natl Canc Inst 2022; 34:43. [DOI: 10.1186/s43046-022-00139-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 07/14/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
EGFR mutation has not been extensively studied in thyroid cancer. This study was conducted to study spectrum of EGFR mutation in thyroid cancer in Kashmiri population for possible therapeutic purpose.
Methods
It was 2 years prospective cross-sectional study conducted at a tertiary care center in which histologically confirmed, untreated thyroid cancers were included. These specimens were subjected to EGFR mutation analysis by AS-PCR method.
Results
There were a total 60 patients with preponderance of females [44(73%) vs 16(27%)]. Most were in the age group of less than 45 years (75%). Most of these patients were non-smokers [50(83.3%) vs 10 (17.3%)]. Papillary thyroid carcinoma (PTC) was the commonest type 48(80%), rest was follicular type (FTC) 12(20%). Well-differentiated carcinoma (WDC) was common than poorly differentiated (PDC) [41(68.4%) vs 19 (31.6%)]. Lymph node metastasis and vascular invasion were present in 32 (53.4%) and 17 (28.4%) respectively. Thirty-two (53.3%) patients were having 15 bp deletion in exon 19 of EGFR. These deletions were common in PTC than FTC, 29(60.5%) vs 3(25%) which was statistically significant (p = 0.04, CI = 0.2). The total mutational rate of T790M in EGFR tyrosine kinase domain (exon 20) was found to be only 8.4% (5 of 60). Only 4 (8.3%) of these mutations were detected in PTC and rest in FTC (1 of 12). Twenty-six (43.3%) of exon 21 were positive for L858R mutation in EGFR tyrosine kinase domain. Married persons and PDC were significant predictors of L858R mutation in EGFR tyrosine kinase domain in thyroid cancer as this was statistically significant in them with p = 0.04, 0.03 respectively.
Conclusion
In our population, PTC is common in females with half of population harboring EGFR mutation and it is statistically significant in poorly differentiated carcinoma and in married individuals.
It implies that EGFR may be used in thyroid cancer as a possible therapeutic agent in our set of population.
Collapse
|
99866
|
Zhu C, Wang S, Du Y, Dai Y, Huai Q, Li X, Du Y, Dai H, Yuan W, Yin S, Wang H. Tumor microenvironment-related gene selenium-binding protein 1 (SELENBP1) is associated with immunotherapy efficacy and survival in colorectal cancer. BMC Gastroenterol 2022; 22:437. [PMID: 36253721 PMCID: PMC9575293 DOI: 10.1186/s12876-022-02532-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 10/07/2022] [Indexed: 11/29/2022] Open
Abstract
Background Selenium-binding protein 1 (SELENBP1), a member of the selenium-containing protein family, plays an important role in malignant tumorigenesis and progression. However, it is currently lacking research about relationship between SELENBP1 and immunotherapy in colorectal cancer (CRC). Methods We first analyzed the expression levels of SELENBP1 based on the Cancer Genome Atlas (TCGA), Oncomine andUALCAN. Chisq.test, Fisher.test, Wilcoxon-Mann-Whitney test and logistic regression were used to analyze the relationship of clinical characteristics with SELENBP1 expression. Then Gene ontology/ Kyoto encyclopedia of genes and genomes (GO/KEGG), Gene set enrichment analysis (GSEA) enrichment analysis to clarify bio-processes and signaling pathways. The cBioPortal was used to perform analysis of mutation sites, types, etc. of SELENBP1. In addition, the correlation of SELENBP1 gene with tumor immune infiltration and prognosis was analyzed using ssGSEA, ESTIMATE, tumor immune dysfunction and rejection (TIDE) algorithm and Kaplan-Meier (KM) Plotter database. Quantitative real-time PCR (qRT-PCR) and western blotting (WB) were used to validate the expression of SELENBP1 in CRC samples and matched normal tissues. Immunohistochemistry (IHC) was further performed to detect the expression of SELENBP1 in CRC samples and matched normal tissues. Results We found that SELENBP1 expression was lower in CRC compared to normal colorectal tissue and was associated with poor prognosis. The aggressiveness of CRC increased with decreased SELENBP1 expression. Enrichment analysis showed that the SELENBP1 gene was significantly enriched in several pathways, such as programmed death 1 (PD-1) signaling, signaling by interleukins, TCR signaling, collagen degradation, costimulation by the CD28 family. Decreased expression of SELENBP1 was associated with DNA methylation and mutation. Immune infiltration analysis identified that SELENBP1 expression was closely related to various immune cells and immune chemokines/receptors. With increasing SELENBP1 expression, immune and stromal components in the tumor microenvironment were significantly decreased. SELENBP1 expression in CRC patients affects patient prognosis by influencing tumor immune infiltration. Beside this, SELENBP1 expression is closely related to the sensitivity of chemotherapy and immunotherapy. Conclusions Survival analysis as well as enrichment and immunoassay results suggest that SELENBP1 can be considered as a promising prognostic biomarker for CRC. SELENBP1 expression is closely associated with immune infiltration and immunotherapy. Collectively, our study provided useful information on the oncogenic role of SELENBP1, contributing to further exploring the underlying mechanisms. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02532-2.
Collapse
Affiliation(s)
- Cheng Zhu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Siya Wang
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230001, China
| | - Yishan Du
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230001, China
| | - Ying Dai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Qian Huai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Xiaolei Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Yingying Du
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Hanren Dai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Wenkang Yuan
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230036, China
| | - Shi Yin
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230001, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China.
| |
Collapse
|
99867
|
Wang M, Bi C, Li H, Lu L, Gao T, Huang P, Liu C, Wang B. The emerging double-edged sword role of Sirtuins in the gastric inflammation-carcinoma sequence revealed by bulk and single-cell transcriptomes. Front Oncol 2022; 12:1004726. [PMID: 36324577 PMCID: PMC9619065 DOI: 10.3389/fonc.2022.1004726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/12/2022] [Indexed: 12/03/2022] Open
Abstract
Histone modification and the inflammation-carcinoma sequence (ICS) have been acknowledgedly implicated in gastric carcinogenesis. However, the extremum expression of some histone modification genes (HMGs) in intestinal metaplasia (IM) rather than GC obscures the roles of HMGs in ICS. In this study, we assumed an explanation that the roles of HMGs in ICS were stage specific. Bulk RNA-seq on endoscopy biopsy samples from a total of 50 patients was accompanied by reanalysis of a set of published single-cell transcriptomes, which cross-sectionally profiled the transcriptomic features of chronic superficial gastritis (SG), atrophy gastritis (AG), IM, and early gastric cancer (GC). Differential analysis observed significantly peaked expression of SIRT6 and SIRT7 at IM. Weighted correlation network analysis on bulk transcriptome recognized significant correlations between SIRT1/6 and IM. The single-cell atlas identified one subgroup of B cells expressing high level of TFF1 (TFF1hi naive B cell) that theoretically played important roles in defending microbial infection, while SIRT6 displayed a positive correlation with TFF1low naive B cells. Moreover, gene set enrichment analysis at different lesions (SG-AG, AG-IM, and IM-GC) highlighted that gene sets contributing to IM, e.g., Brush Border, were largely enriched from co-expressing genes of Sirtuins (SIRTs) in AG-IM. Surveys of the genes negatively correlated with SIRT6 in public databases considered SIRT6 as tumor suppressors, which was confirmed by the cell proliferation and migration assays after transient transfection of SIRT6 overexpression vector into AGS cells. All the above observations were then confirmed by serial section-based immunohistochemistry against Ki-67, MUC2, MUC5AC, p53, and SIRT6 on the endoscopic submucosal dissection tissue. By contrast, the expression of the other HMGs varied even opposite within same family. Taken together, this study preliminarily demonstrated the two-edged sword role of SIRTs in ICS and, by extension, showed that the roles of HMGs in ICS were probably stage specific. Our study may provide new insights into and attract attention on gastric prevention and therapy targeting HMGs.
Collapse
Affiliation(s)
- Mengyang Wang
- Department of Immunology, Binzhou Medical University, Yantai, China
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Chenxiao Bi
- Department of Immunology, Binzhou Medical University, Yantai, China
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Hong Li
- Department of Pathology, Binzhou Medical University Hospital, Binzhou, China
| | - Lizhen Lu
- Department of Pathology, Binzhou Medical University Hospital, Binzhou, China
| | - Tao Gao
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Panpan Huang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Chengxia Liu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- *Correspondence: Chengxia Liu, ; Bin Wang,
| | - Bin Wang
- Department of Immunology, Binzhou Medical University, Yantai, China
- *Correspondence: Chengxia Liu, ; Bin Wang,
| |
Collapse
|
99868
|
Hu B, Zou T, Qin W, Shen X, Su Y, Li J, Chen Y, Zhang Z, Sun H, Zheng Y, Wang CQ, Wang Z, Li TE, Wang S, Zhu L, Wang X, Fu Y, Ren X, Dong Q, Qin LX. Inhibition of EGFR Overcomes Acquired Lenvatinib Resistance Driven by STAT3-ABCB1 Signaling in Hepatocellular Carcinoma. Cancer Res 2022; 82:3845-3857. [PMID: 36066408 PMCID: PMC9574378 DOI: 10.1158/0008-5472.can-21-4140] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 07/18/2022] [Accepted: 08/29/2022] [Indexed: 01/07/2023]
Abstract
UNLABELLED Lenvatinib is an inhibitor of multiple receptor tyrosine kinases that was recently authorized for first-line treatment of hepatocellular carcinoma (HCC). However, the clinical benefits derived from lenvatinib are limited, highlighting the urgent need to understand mechanisms of resistance. We report here that HCC cells develop resistance to lenvatinib by activating EGFR and stimulating the EGFR-STAT3-ABCB1 axis. Lenvatinib resistance was accompanied by aberrant cholesterol metabolism and lipid raft activation. ABCB1 was activated by EGFR in a lipid raft-dependent manner, which significantly enhanced the exocytosis of lenvatinib to mediate resistance. Furthermore, clinical specimens of HCC showed a correlation between the activation of the EGFR-STAT3-ABCB1 pathway and lenvatinib response. Erlotinib, an EGFR inhibitor that has also been shown to inhibit ABCB1, suppressed lenvatinib exocytosis, and combined treatment with lenvatinib and erlotinib demonstrated a significant synergistic effect on HCC both in vitro and in vivo. Taken together, these findings characterize a mechanism of resistance to a first-line treatment for HCC and offer a practical means to circumvent resistance and treat the disease. SIGNIFICANCE HCC cells acquire resistance to lenvatinib by activating the EGFR-STAT3-ABCB1 pathway, identifying combined treatment with erlotinib as a strategy to overcome acquired resistance and improve the clinical benefit of lenvatinib.
Collapse
Affiliation(s)
- Beiyuan Hu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tiantian Zou
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wei Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiaotian Shen
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yinghan Su
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jianhua Li
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Yang Chen
- Phase I Clinical Trial Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ze Zhang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Haoting Sun
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yan Zheng
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chao-Qun Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhengxin Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Tian-En Li
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shun Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Le Zhu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xufeng Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yan Fu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xudong Ren
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qiongzhu Dong
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
99869
|
Mao R, Kong W, He Y. The affinity of antigen-binding domain on the antitumor efficacy of CAR T cells: Moderate is better. Front Immunol 2022; 13:1032403. [PMID: 36325345 PMCID: PMC9618871 DOI: 10.3389/fimmu.2022.1032403] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
The overall efficacy of chimeric antigen receptor modified T cells (CARTs) remain limited in solid tumors despite intensive studies that aim at targeting multiple antigens, enhancing migration, reducing tonic signaling, and improving tumor microenvironment. On the other hand, how the affinity and engaging kinetics of antigen-binding domain (ABD) affects the CART's efficacy has not been carefully investigated. In this article, we first analyzed 38 published solid tumor CART trials and correlated the response rate to their ABD affinity. Not surprisingly, majority (25 trials) of the CARTs utilized high-affinity ABDs, but generated merely 5.7% response rate. In contrast, 35% of the patients treated with the CARTs built from moderate-affinity ABDs had clinical responses. Thus, CARTs with moderate-affinity ABDs not only have less off-target toxicity, but also are more effective. We then reviewed the effects of ABD affinity on the biology and function of CARTs, providing further evidence that moderate-affinity ABDs may be better in CART development. In the end, we propose that a fast-on/fast-off (high Kon and Koff ) kinetics of CART-target engagement in solid tumor allow CARTs to generate sufficient signaling to kill tumor cells without being driven to exhaustion. We believe that studying the ABD affinity and the kinetics of CART-tumor interaction may hold a key to designing effective CARTs for solid tumors.
Collapse
Affiliation(s)
- Rui Mao
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Wanqing Kong
- South Carolina Governors School for Science and Math, Hartsville, SC, United States
| | - Yukai He
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
99870
|
Wang C, Lin R, Yao S. Recent Advances in 18F-Labeled Amino Acids Synthesis and Application. Pharmaceutics 2022; 14:pharmaceutics14102207. [PMID: 36297641 PMCID: PMC9609324 DOI: 10.3390/pharmaceutics14102207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Radiolabeled amino acids are an important class of agents for positron emission tomography imaging that target amino acid transporters in many tumor types. Traditional 18F-labeled amino acid synthesis strategies are always based on nucleophilic aromatic substitution reactions with multistep radiosynthesis and low radiochemical yields. In recent years, new 18F-labeling methodologies such as metal-catalyzed radiofluorination and heteroatom (B, P, S, Si, etc.)-18F bond formation are being effectively used to synthesize radiopharmaceuticals. This review focuses on recent advances in the synthesis, radiolabeling, and application of a series of 18F-labeled amino acid analogs using new 18F-labeling strategies.
Collapse
|
99871
|
Brackmann LK, Foraita R, Schwarz H, Galetzka D, Zahnreich S, Hankeln T, Löbrich M, Poplawski A, Grabow D, Blettner M, Schmidberger H, Marron M. Late health effects and changes in lifestyle factors after cancer in childhood with and without subsequent second primary cancers – the KiKme case-control study. Front Oncol 2022; 12:1037276. [PMID: 36324589 PMCID: PMC9618813 DOI: 10.3389/fonc.2022.1037276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022] Open
Abstract
Background Improved treatments for childhood cancer result in a growing number of long-term childhood cancer survivors (CCS). The diagnosis and the prevalence of comorbidities may, however, influence their lifestyle later in life. Nonetheless, little is known about differences in late effects between CCS of a first primary neoplasm (FPN) in childhood and subsequent second primary neoplasms (SPN) and their impact on lifestyle. Therefore, we aim to investigate associations between the occurrence of FPN or SPN and various diseases and lifestyle in the later life of CCS. Methods CCS of SPN (n=101) or FPN (n=340) and cancer-free controls (n=150) were matched by age and sex, and CCS additionally by year and entity of FPN. All participants completed a self-administered questionnaire on anthropometric and socio-economic factors, medical history, health status, and lifestyle. Mean time between FPN diagnosis and interview was 27.3 years for SPN and 26.2 years for FPN CCS. To confirm results from others and to generate new hypotheses on late effects of childhood cancer as well as CCS´ lifestyles, generalized linear mixed models were applied. Results CCS were found to suffer more likely from diseases compared to cancer-free controls. In detail, associations with cancer status were observed for hypercholesterinemia and thyroid diseases. Moreover, CCS were more likely to take regular medication compared to controls. A similar association was observed for CCS of SPN compared to CCS of FPN. In contrast to controls, CCS rarely exercise more than 5 hours per week, consumed fewer soft and alcoholic drinks, and were less likely to be current, former, or passive smokers. Additionally, they were less likely overweight or obese. All other exploratory analyses performed on cardiovascular, chronic lung, inflammatory bone, allergic, and infectious diseases, as well as on a calculated health-score revealed no association with tumor status. Conclusion CCS were more affected by pathologic conditions and may consequently take more medication, particularly among CCS of SPN. The observed higher disease burden is likely related to the received cancer therapy. To reduce the burden of long-term adverse health effects in CCS, improving cancer therapies should therefore be in focus of research in this area.
Collapse
Affiliation(s)
- Lara Kim Brackmann
- Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology – BIPS, Bremen, Germany
- Faculty of Mathematics and Computer Science, University of Bremen, Bremen, Germany
| | - Ronja Foraita
- Biometry and Data Management, Leibniz Institute for Prevention Research and Epidemiology – BIPS, Bremen, Germany
| | - Heike Schwarz
- Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology – BIPS, Bremen, Germany
| | - Danuta Galetzka
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Sebastian Zahnreich
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas Hankeln
- Institute of Organismic and Molecular Evolution, Molecular Genetics and Genome Analysis, Johannes Gutenberg University, Mainz, Germany
| | - Markus Löbrich
- Radiation Biology and deoxyribonucleic acid (DNA) Repair, Technical University of Darmstadt, Darmstadt, Germany
| | - Alicia Poplawski
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Desiree Grabow
- German Childhood Cancer Registry, Institute for Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Maria Blettner
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Heinz Schmidberger
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Manuela Marron
- Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology – BIPS, Bremen, Germany
- *Correspondence: Manuela Marron,
| |
Collapse
|
99872
|
He S, Gu X, Yang J, Xu F, Hu J, Wang W, Huang Y, Lou B, Ding T, Zhou L, Ye D, Yu K, Dong J. Sphingomyelin synthase 2 is a positive regulator of the CSF1R-STAT3 pathway in pancreatic cancer-associated macrophage. Front Pharmacol 2022; 13:902016. [PMID: 36324684 PMCID: PMC9618885 DOI: 10.3389/fphar.2022.902016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/22/2022] [Indexed: 04/07/2025] Open
Abstract
Background: Tumor-associated macrophages (TAMs) are one of the most abundant immune cells in the pancreatic cancer stroma and are related to the poor prognosis of pancreatic ductal adenocarcinoma (PDAC) patients. Therefore, targeting tumor-associated macrophages is a possible strategy for the treatment of pancreatic cancer. Purpose: We would like to investigate the role of sphingomyelin synthase 2 (SMS2) and the effect of the synthase 2 selective inhibitor YE2 in TAMs and the pancreatic tumor microenvironment. In addition, we also would like to investigate the mechanism by which YE2 attenuates macrophage M2 polarization. Methods: YE2 was utilized to treat macrophages (in vitro) and mice (in vivo). Western blotting and real-time PCR were used to detect the protein levels and mRNA levels of macrophage M2 polarization markers and their downstream signaling pathways. Sphingomyelin synthase 2 gene knockout (KO) mice and their controls were used to establish a PANC-02 orthotopic pancreatic cancer model, and immune cell infiltration in the tumor tissue was analyzed by immunohistochemistry (IHC). Results: We found that sphingomyelin synthase 2 mRNA expression is positively correlated with tumor-associated macrophages, the immunosuppressive microenvironment, and poor prognosis in pancreatic ductal adenocarcinoma patients. Sphingomyelin synthase 2 deficiency was confirmed to have an inhibitory effect on the growth of orthotopic PANC-02 tumors in vivo. The deficiency not only reduced the infiltration of tumor-associated macrophages but also regulated other immune components in the tumor microenvironment. In tissue culture, YE2 inhibited M2 polarization in both bone marrow-derived macrophages (BMDMs) and THP-1 macrophages and eliminated the protumor effect of M2 macrophages. In the mouse model, YE2 treatment reduced the infiltration of TAMs and regulated other immune components in the tumor microenvironment, slowing the progression of PANC-02 tumors. In terms of mechanism, we found that the inhibition of sphingomyelin synthase 2 could downregulate the expression of IL4Rα and CSF1R, thereby attenuating M2 polarization. Conclusion: The sphingomyelin synthase 2 inhibitor YE2 or sphingomyelin synthase 2 deficiency can prevent macrophage M2 polarization in pancreatic cancer, and sphingomyelin synthase 2 could be a new potential target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Shuhua He
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiang Gu
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Jintong Yang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Fei Xu
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiachun Hu
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Yiheng Huang
- Department of Clinical Medicine, Shanghai Jiaotong University of Medicine, Shanghai, China
| | - Bin Lou
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Tingbo Ding
- Experiment & Teaching Center, School of Pharmacy, Fudan University, Shanghai, China
| | - Lu Zhou
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Deyong Ye
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Ker Yu
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Jibin Dong
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai, China
| |
Collapse
|
99873
|
Minciuna I, van Kleef LA, Stefanescu H, Procopet B. Is Fasting Good When One Is at Risk of Liver Cancer? Cancers (Basel) 2022; 14:5084. [PMID: 36291868 PMCID: PMC9600146 DOI: 10.3390/cancers14205084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC), one of the leading causes of cancer-related deaths worldwide, is a multistep process that usually develops in the background of cirrhosis, but also in a non-cirrhotic state in patients with non-alcoholic fatty liver disease (NAFLD) or viral hepatis. Emerging evidence suggests that intermittent fasting can reduce the risk of cancer development and could improve response and tolerance to treatment through the metabolic and hormonal adaptations induced by the low energy availability that finally impairs cancer cells' adaptability, survival and growth. The current review will outline the beneficial effects of fasting in NAFLD/NASH patients and the possible mechanisms that can prevent HCC development, including circadian clock re-synchronization, with a special focus on the possibility of applying this dietary intervention to cirrhotic patients.
Collapse
Affiliation(s)
- Iulia Minciuna
- Regional Institute of Gastroenterology and Hepatology Octavian Fodor, 400394 Cluj-Napoca, Romania
- 3rd Medical Department, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj-Napoca, Romania
| | - Laurens A. van Kleef
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Horia Stefanescu
- Regional Institute of Gastroenterology and Hepatology Octavian Fodor, 400394 Cluj-Napoca, Romania
| | - Bogdan Procopet
- Regional Institute of Gastroenterology and Hepatology Octavian Fodor, 400394 Cluj-Napoca, Romania
- 3rd Medical Department, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj-Napoca, Romania
| |
Collapse
|
99874
|
Zhou RW, Xu J, Martin TC, Zachem AL, He J, Ozturk S, Demircioglu D, Bansal A, Trotta AP, Giotti B, Gryder B, Shen Y, Wu X, Carcamo S, Bosch K, Hopkins B, Tsankov A, Steinhagen R, Jones DR, Asara J, Chipuk JE, Brody R, Itzkowitz S, Chio IIC, Hasson D, Bernstein E, Parsons RE. A local tumor microenvironment acquired super-enhancer induces an oncogenic driver in colorectal carcinoma. Nat Commun 2022; 13:6041. [PMID: 36253360 PMCID: PMC9576746 DOI: 10.1038/s41467-022-33377-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022] Open
Abstract
Tumors exhibit enhancer reprogramming compared to normal tissue. The etiology is largely attributed to cell-intrinsic genomic alterations. Here, using freshly resected primary CRC tumors and patient-matched adjacent normal colon, we find divergent epigenetic landscapes between CRC tumors and cell lines. Intriguingly, this phenomenon extends to highly recurrent aberrant super-enhancers gained in CRC over normal. We find one such super-enhancer activated in epithelial cancer cells due to surrounding inflammation in the tumor microenvironment. We restore this super-enhancer and its expressed gene, PDZK1IP1, following treatment with cytokines or xenotransplantation into nude mice, thus demonstrating cell-extrinsic etiology. We demonstrate mechanistically that PDZK1IP1 enhances the reductive capacity CRC cancer cells via the pentose phosphate pathway. We show this activation enables efficient growth under oxidative conditions, challenging the previous notion that PDZK1IP1 acts as a tumor suppressor in CRC. Collectively, these observations highlight the significance of epigenomic profiling on primary specimens.
Collapse
Affiliation(s)
- Royce W Zhou
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jia Xu
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tiphaine C Martin
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexis L Zachem
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - John He
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sait Ozturk
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Deniz Demircioglu
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ankita Bansal
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrew P Trotta
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bruno Giotti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Berkley Gryder
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Yao Shen
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Xuewei Wu
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Saul Carcamo
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kaitlyn Bosch
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Benjamin Hopkins
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexander Tsankov
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Randolph Steinhagen
- Division of Colon and Rectal Surgery, Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Drew R Jones
- Metabolomics Core Resource Laboratory, NYU Langone Health, New York, NY, 10016, USA
| | - John Asara
- Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rachel Brody
- Mount Sinai Biorepository, Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Steven Itzkowitz
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Iok In Christine Chio
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
| | - Dan Hasson
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Emily Bernstein
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ramon E Parsons
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
99875
|
Li Y, Xin X, Song W, Zhang X, Chen S, Wang Q, Li A, Li Y. VHL syndrome without clear family history: A rare case report and literature review of Chinese patients. Front Neurol 2022; 13:951054. [PMID: 36324386 PMCID: PMC9618664 DOI: 10.3389/fneur.2022.951054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/05/2022] [Indexed: 11/26/2022] Open
Abstract
Objective To analyze the clinical manifestations and imaging features of a hospitalized patient with intermittent headache who was finally diagnosed with von Hippel–Lindau (VHL) syndrome and to perform whole-exon gene detection to improve the understanding of the diagnosis and treatment strategies of the disease. Methods A case of suspected VHL syndrome in Shanxi Provincial People's Hospital was analyzed. Proband DNA was also extracted for whole exome sequencing and screened for causative mutation sites, which were validated by Sanger sequencing. The literature about VHL gene mutations in Chinese patients in the past 10 years were also reviewed. Results There is a heterozygous mutation site c.499C > G on the VHL gene on the short arm of chromosome 3 of the patient, which is a missense mutation. The mutation results in the substitution of arginine with glycine at amino acid 167 of the encoded protein, which may be primarily responsible for the disease in the patient with VHL syndrome. However, the mutation did not occur in other family members. Conclusion Early recognition and treatment of VHL syndrome can be available with genetic testing technology. Strengthening the understanding of this complex genetic disease and improving the diagnostic rate of VHL syndrome are helpful for the precise treatment of patients with this disease, which may help prolong the survival time of patients to a certain extent and improve their quality of life.
Collapse
Affiliation(s)
- Yaheng Li
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, China
| | - Xiaohong Xin
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi University, Taiyuan, China
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan, China
| | - Wenzhu Song
- School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Xuan Zhang
- Department of Neurosurgery, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Shengli Chen
- Department of Neurosurgery, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Qian Wang
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, China
| | - Aizhong Li
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, China
| | - Yafeng Li
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, China
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi University, Taiyuan, China
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan, China
- *Correspondence: Yafeng Li
| |
Collapse
|
99876
|
Zhou Y, Li ZL, Ding L, Zhang XJ, Liu NC, Liu SS, Wang YF, Ma RX. Long noncoding RNA SNHG5 promotes podocyte injury via the microRNA-26a-5p/TRPC6 pathway in diabetic nephropathy. J Biol Chem 2022; 298:102605. [PMID: 36257404 PMCID: PMC9694110 DOI: 10.1016/j.jbc.2022.102605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022] Open
Abstract
Podocyte injury is a characteristic pathological hallmark of diabetic nephropathy (DN). However, the exact mechanism of podocyte injury in DN is incompletely understood. This study was conducted using db/db mice and immortalized mouse podocytes. High-throughput sequencing was used to identify the differentially expressed long noncoding RNAs in kidney of db/db mice. The lentiviral shRNA directed against long noncoding RNA small nucleolar RNA host gene 5 (SNHG5) or microRNA-26a-5p (miR-26a-5p) agomir was used to treat db/db mice to regulate the SNHG5/miR-26a-5p pathway. Here, we found that the expression of transient receptor potential canonical type 6 (TRPC6) was significantly increased in injured podocytes under the condition of DN, which was associated with markedly decreased miR-26a-5p. We determined that miR-26a-5p overexpression ameliorated podocyte injury in DN via binding to 3'-UTR of Trpc6, as evidenced by the markedly reduced activity of luciferase reporters by miR-26a-5p mimic. Then, the upregulated SNHG5 in podocytes and kidney in DN was identified, and it was proved to sponge to miR-26a-5p directly using luciferase activity, RNA immunoprecipitation, and RNA pull-down assay. Knockdown of SNHG5 attenuated podocyte injury in vitro, accompanied by an increased expression of miR-26a-5p and decreased expression of TRPC6, demonstrating that SNHG5 promoted podocyte injury by controlling the miR-26a-5p/TRPC6 pathway. Moreover, knockdown of SNHG5 protects against podocyte injury and progression of DN in vivo. In conclusion, SNHG5 promotes podocyte injury via the miR-26a-5p/TRPC6 pathway in DN. Our findings provide novel insights into the pathophysiology of podocyte injury and a potential new therapeutic strategy for DN.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Lin Ding
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xing-Jian Zhang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Nan-Chi Liu
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shan-Shan Liu
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan-Fei Wang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Rui-Xia Ma
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China,For correspondence: Rui-Xia Ma
| |
Collapse
|
99877
|
Gao X, Wang C, Abdelrahman S, Kady N, Murga-Zamalloa C, Gann P, Sverdlov M, Wolfe A, Polk A, Brown N, Bailey NG, Inamdar K, Casavilca S, Montes J, Barrionuevo C, Taxa L, Reneau J, Siebel CW, Maillard I, Wilcox RA. Notch Signaling Promotes Mature T-Cell Lymphomagenesis. Cancer Res 2022; 82:3763-3773. [PMID: 36006995 PMCID: PMC9588752 DOI: 10.1158/0008-5472.can-22-1215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/15/2022] [Accepted: 08/22/2022] [Indexed: 01/26/2023]
Abstract
Peripheral T-cell lymphomas (PTCL) are agressive lymphomas that develop from mature T cells. The most common PTCLs are genetically, molecularly, and clinically diverse and are generally associated with dismal outcomes. While Notch signaling plays a critically important role in both the development of immature T cells and their malignant transformation, its role in PTCL is poorly understood, despite the increasingly appreciated function of Notch in regulating the proliferation and differentiation of mature T cells. Here, we demonstrate that Notch receptors and their Delta-like family ligands (DLL1/DLL4) play a pathogenic role in PTCL. Notch1 activation was observed in common PTCL subtypes, including PTCL-not otherwise specified (NOS). In a large cohort of PTCL-NOS biopsies, Notch1 activation was significantly associated with surrogate markers of proliferation. Complementary genetically engineered mouse models and spontaneous PTCL models were used to functionally examine the role of Notch signaling, and Notch1/Notch2 blockade and pan-Notch blockade using dominant-negative MAML significantly impaired the proliferation of malignant T cells and PTCL progression in these models. Treatment with DLL1/DLL4 blocking antibodies established that Notch signaling is ligand-dependent. Together, these findings reveal a role for ligand-dependent Notch signaling in driving peripheral T-cell lymphomagenesis. SIGNIFICANCE This work demonstrates that ligand-dependent Notch activation promotes the growth and proliferation of mature T-cell lymphomas, providing new therapeutic strategies for this group of aggressive lymphomas.
Collapse
Affiliation(s)
- Xin Gao
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Chenguang Wang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Suhaib Abdelrahman
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Nermin Kady
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | | | - Peter Gann
- Department of Pathology, University of Illinois Chicago, Chicago, IL
| | - Maria Sverdlov
- Department of Pathology, University of Illinois Chicago, Chicago, IL
| | - Ashley Wolfe
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Avery Polk
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Noah Brown
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | | | - Kedar Inamdar
- Department of Pathology, Henry Ford Hospital, Detroit, MI
| | - Sandro Casavilca
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas (INEN), Lima, Peru
| | - Jaime Montes
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas (INEN), Lima, Peru
| | - Carlos Barrionuevo
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas (INEN), Lima, Peru
| | - Luis Taxa
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas (INEN), Lima, Peru
| | - John Reneau
- Department of Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | - Ivan Maillard
- Department of Medicine, Division of Hematology/Oncology, University of Pennsylvania, Philadelphia, PA
| | - Ryan A. Wilcox
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
99878
|
Garcia-Melo LF, Morales-Rodríguez M, Madrigal-Bujaidar E, Madrigal-Santillán EO, Morales-González JA, Pineda Cruces RN, Campoy Ramírez JA, Damian-Matsumura P, Tellez-Plancarte A, Batina N, Álvarez-González I. Development of a Nanostructured Electrochemical Genosensor for the Detection of the K-ras Gene. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2022; 2022:6575140. [PMID: 36299712 PMCID: PMC9592225 DOI: 10.1155/2022/6575140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/06/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
In the scientific literature, it has been documented that electrochemical genosensors are novel analytical tools with proven clinical diagnostic potential for the identification of carcinogenic processes due to genetic and epigenetic alterations, as well as infectious diseases due to viruses or bacteria. In the present work, we describe the construction of an electrochemical genosensor for the identification of the k12p.1 mutation; it was based on use of Screen-Printed Gold Electrode (SPGE), Cyclic Voltammetry (CV), and Atomic Force Microscopy (AFM), for the monitoring the electron transfer trough the functionalized nanostructured surface and corresponding morphological changes. The sensitivity of the genosensor showed a linear response for the identification of the k12p.1 mutation of the K-ras gene in the concentration range of 10 fM to 1 μM with a detection limit of 7.96 fM in the presence of doxorubicin (Dox) as DNA intercalating agent and indicator of the hybridization reaction. Thus, the electrochemical genosensor developed could be useful for the identification of diseases related with the K-ras oncogene.
Collapse
Affiliation(s)
- Luis Fernando Garcia-Melo
- Division de Ingeniería en Nanotecnología, Universidad Politécnica del Valle de México, Av. Mexiquense s/n esquina Av. Universidad Politécnica, Tultitlan Estado de México, CP 54910, Mexico
- Laboratorio de Nanotecnología e Ingeniería Molecular Área Electroquímica, Departamento de Química, CBI, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), Av. San Rafael Atlixco 186, Iztapalapa, CP 09340, México City, Mexico
| | - Miguel Morales-Rodríguez
- Division de Ingeniería en Nanotecnología, Universidad Politécnica del Valle de México, Av. Mexiquense s/n esquina Av. Universidad Politécnica, Tultitlan Estado de México, CP 54910, Mexico
| | - Eduardo Madrigal-Bujaidar
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Avenida Wilfrido Massieu s/n Col. Zacatenco Del. Gustavo A. Madero, CP 07738, Ciudad de México, Mexico
| | - Eduardo O. Madrigal-Santillán
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Unidad Casco de Santo Tomás, Plan de San Luis y Díaz Mirón, Ciudad de México, CP 11340, Mexico
| | - José Antonio Morales-González
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Unidad Casco de Santo Tomás, Plan de San Luis y Díaz Mirón, Ciudad de México, CP 11340, Mexico
| | - Rosa Natali Pineda Cruces
- Laboratorio de Nanotecnología e Ingeniería Molecular Área Electroquímica, Departamento de Química, CBI, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), Av. San Rafael Atlixco 186, Iztapalapa, CP 09340, México City, Mexico
| | - Jorge Alfredo Campoy Ramírez
- Laboratorio de Nanotecnología e Ingeniería Molecular Área Electroquímica, Departamento de Química, CBI, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), Av. San Rafael Atlixco 186, Iztapalapa, CP 09340, México City, Mexico
| | - Pablo Damian-Matsumura
- Laboratorio de Endocrinología Molecular, Departamento de Biología de la Reproducción, CBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), México City, Mexico
| | - Alexandro Tellez-Plancarte
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Avenida Wilfrido Massieu s/n Col. Zacatenco Del. Gustavo A. Madero, CP 07738, Ciudad de México, Mexico
| | - Nikola Batina
- Laboratorio de Nanotecnología e Ingeniería Molecular Área Electroquímica, Departamento de Química, CBI, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), Av. San Rafael Atlixco 186, Iztapalapa, CP 09340, México City, Mexico
| | - Isela Álvarez-González
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Avenida Wilfrido Massieu s/n Col. Zacatenco Del. Gustavo A. Madero, CP 07738, Ciudad de México, Mexico
| |
Collapse
|
99879
|
Yan B. Actuators for Implantable Devices: A Broad View. MICROMACHINES 2022; 13:1756. [PMID: 36296109 PMCID: PMC9610948 DOI: 10.3390/mi13101756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/12/2022] [Accepted: 10/06/2022] [Indexed: 06/16/2023]
Abstract
The choice of actuators dictates how an implantable biomedical device moves. Specifically, the concept of implantable robots consists of the three pillars: actuators, sensors, and powering. Robotic devices that require active motion are driven by a biocompatible actuator. Depending on the actuating mechanism, different types of actuators vary remarkably in strain/stress output, frequency, power consumption, and durability. Most reviews to date focus on specific type of actuating mechanism (electric, photonic, electrothermal, etc.) for biomedical applications. With a rapidly expanding library of novel actuators, however, the granular boundaries between subcategories turns the selection of actuators a laborious task, which can be particularly time-consuming to those unfamiliar with actuation. To offer a broad view, this study (1) showcases the recent advances in various types of actuating technologies that can be potentially implemented in vivo, (2) outlines technical advantages and the limitations of each type, and (3) provides use-specific suggestions on actuator choice for applications such as drug delivery, cardiovascular, and endoscopy implants.
Collapse
Affiliation(s)
- Bingxi Yan
- Department of Electrical and Computer Engineering, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
99880
|
Altwegg KA, Viswanadhapalli S, Mann M, Chakravarty D, Krishnan S, Liu Z, Liu J, Pratap UP, Ebrahimi B, Sanchez JR, Li X, Ma S, Park BH, Santhamma B, Chen Y, Lai Z, Raj GV, Yuan Y, Zhou D, Sareddy GR, Tekmal RR, McHardy S, Huang THM, Rao MK, Vankayalapati H, Vadlamudi RK. A First-in-Class Inhibitor of ER Coregulator PELP1 Targets ER+ Breast Cancer. Cancer Res 2022; 82:3830-3844. [PMID: 35950923 PMCID: PMC9588738 DOI: 10.1158/0008-5472.can-22-0698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/21/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022]
Abstract
Most patients with estrogen receptor alpha-positive (ER+) breast cancers initially respond to treatment but eventually develop therapy resistance with disease progression. Overexpression of oncogenic ER coregulators, including proline, glutamic acid, and leucine-rich protein 1 (PELP1), are implicated in breast cancer progression. The lack of small molecules that inhibits PELP1 represents a major knowledge gap. Here, using a yeast-two-hybrid screen, we identified novel peptide inhibitors of PELP1 (PIP). Biochemical assays demonstrated that one of these peptides, PIP1, directly interacted with PELP1 to block PELP1 oncogenic functions. Computational modeling of PIP1 revealed key residues contributing to its activity and facilitated the development of a small-molecule inhibitor of PELP1, SMIP34, and further analyses confirmed that SMIP34 directly bound to PELP1. In breast cancer cells, SMIP34 reduced cell growth in a dose-dependent manner. SMIP34 inhibited proliferation of not only wild-type (WT) but also mutant (MT) ER+ and therapy-resistant breast cancer cells, in part by inducing PELP1 degradation via the proteasome pathway. RNA sequencing analyses showed that SMIP34 treatment altered the expression of genes associated with estrogen response, cell cycle, and apoptosis pathways. In cell line-derived and patient-derived xenografts of both WT and MT ER+ breast cancer models, SMIP34 reduced proliferation and significantly suppressed tumor progression. Collectively, these results demonstrate SMIP34 as a first-in-class inhibitor of oncogenic PELP1 signaling in advanced breast cancer. SIGNIFICANCE Development of a novel inhibitor of oncogenic PELP1 provides potential therapeutic avenues for treating therapy-resistant, advanced ER+ breast cancer.
Collapse
Affiliation(s)
- Kristin A. Altwegg
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX 78229
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX 78229
| | - Monica Mann
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
| | | | - Samaya Krishnan
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
| | - Zexuan Liu
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
- Department of Oncology, Xiangya Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R. China
| | - Junhao Liu
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
- Department of Oncology, Xiangya Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R. China
| | - Uday P. Pratap
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX 78229
| | - Behnam Ebrahimi
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX 78229
| | - John R. Sanchez
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
| | - Xiaonan Li
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
| | - Shihong Ma
- Department of Urology, UT Southwestern Medical Center, Dallas, TX
| | - Ben H. Park
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | | | - Yidong Chen
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX 78229
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229
| | - Zhao Lai
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX 78229
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229
| | - Ganesh V. Raj
- Department of Urology, UT Southwestern Medical Center, Dallas, TX
| | - Yaxia Yuan
- Department of Biochemistry and Structural Biology, and Center for Innovative Drug Discovery, UT Health San Antonio, San Antonio, TX 78229
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology, and Center for Innovative Drug Discovery, UT Health San Antonio, San Antonio, TX 78229
| | - Gangadhara R. Sareddy
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX 78229
| | - Rajeshwar R. Tekmal
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX 78229
| | - Stan McHardy
- Department of Chemistry, University of Texas San Antonio, San Antonio, Texas, USA
| | - Tim H. -M. Huang
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX 78229
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX 78229
| | - Manjeet K. Rao
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX 78229
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229
| | | | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX 78229
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX 78229
- Audie L. Murphy South Texas Veterans Health Care System, San Antonio, TX
| |
Collapse
|
99881
|
Jiang W, Li X, Xiang C, Zhou W. Neutrophils in pancreatic cancer: Potential therapeutic targets. Front Oncol 2022; 12:1025805. [PMID: 36324574 PMCID: PMC9618950 DOI: 10.3389/fonc.2022.1025805] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/28/2022] [Indexed: 08/30/2023] Open
Abstract
Pancreatic cancer is a digestive system malignancy and poses a high mortality worldwide. Traditionally, neutrophils have been thought to play a role in acute inflammation. In contrast, their importance during tumor diseases has been less well studied. Generally, neutrophils are recruited into the tumor microenvironment and exert inflammation and tumor-promoting effects. As an essential part of the tumor microenvironment, neutrophils play diverse roles in pancreatic cancer, such as angiogenesis, progression, metastasis and immunosuppression. Additionally, neutrophils can be a new potential therapeutic target in cancer. Inhibitors of cytokines, chemokines and neutrophil extracellular traps can exert antitumor effects. In this review, we describe the role of neutrophils in the development and progression of pancreatic cancer, discuss their potential as therapeutic targets, and aim to provide ideas for improving the prognosis of patients with this malignant tumor disease.
Collapse
Affiliation(s)
- Wenkai Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xin Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Caifei Xiang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
99882
|
Cai Y, Lin J, Wang Z, Ma Y, Pan J, Liu Y, Zhao Z. Identification and validation of a lipid metabolism gene signature for predicting biochemical recurrence of prostate cancer after radical prostatectomy. Front Oncol 2022; 12:1009921. [PMID: 36324578 PMCID: PMC9619088 DOI: 10.3389/fonc.2022.1009921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/03/2022] [Indexed: 12/02/2022] Open
Abstract
Background Pro5state cancer is one of the most commonly diagnosed cancers in men worldwide and biochemical recurrence occurs in approximately 25% of patients after radical prostatectomy. Current decisions regarding biochemical recurrence after radical prostatectomy are largely dependent on clinicopathological parameters, which are less accurate. A growing body of research suggests that lipid metabolism influences tumor development and treatment, and that prostate cancer is not only a malignancy but also a lipid metabolism disease. Therefore, this study aimed to identify the prognostic value of lipid metabolism-related gene signaling disease to better predict biochemical recurrence and contribute to clinical decision-making. Methods Expression data and corresponding clinical information were obtained from The Cancer Genome Atlas (TCGA) database and the MSKCC database. Candidate modules closely associated with BCR were screened by univariate and LASSOcox regression analyses, and multivariate Cox regression analyses were performed to construct gene signatures. Kaplan-Meier (KM) survival analysis, time-dependent subject operating curves (ROC), independent prognostic analysis, and Nomogram were also used to assess the prognostic value of the signatures. In addition, Gene Ontology Analysis (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were used to explore potential biological pathways. Results A 6-gene lipid metabolism-related gene signature was successfully constructed and validated to predict biochemical recurrence in prostate cancer patients. In addition, we identified the 6-gene signature as an independent risk factor. Functional analysis showed that lipid metabolism-related genes were closely associated with arachidonic acid metabolism, PPAR transduction signaling pathway, fatty acid metabolism, peroxisome, and glycerophospholipid metabolism. Prognostic models were associated with immune cell infiltration. Conclusion We have successfully developed a novel lipid metabolism-related gene signature that is highly effective in predicting BCR in patients with limited prostate cancer after RP and created a prognostic Nomogram. Furthermore, the signature may help clinicians to select high-risk subpopulations, predict patient survival, and facilitate more personalized treatment than traditional clinical factors.
Collapse
|
99883
|
Baseline anemia predicts a poor prognosis in patients with non-small cell lung cancer with epidermal growth factor receptor mutations: a retrospective study. BMC Pulm Med 2022; 22:381. [PMID: 36253772 PMCID: PMC9575199 DOI: 10.1186/s12890-022-02158-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/13/2022] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Anemia is relatively common in cancer patients, and baseline anemia is associated with poor survival in patients with non-small cell lung cancer (NSCLC). However, there is a lack of large-sample studies of patients with NSCLC with epidermal growth factor receptor (EGFR) mutations. METHODS We retrospectively analyzed anemia‑related data for patients with NSCLC and EGFR mutations who were admitted to Zhejiang Cancer Hospital from January 2013 to June 2019 and treated with targeted therapy. The patients' clinicopathological features were evaluated by χ2 tests and the relationships between clinical characteristics and prognosis were investigated using Kaplan-Meier and multivariate Cox regression analyses. RESULTS A total of 2,029 patients treated with EGFR-tyrosine kinase inhibitors (TKIs) were finally enrolled in this study, of whom 24.6% had baseline anemia. Patients without baseline anemia had longer median overall survival (OS) than patients with baseline anemia (36.10 vs. 29.10 months, P = 0.001), and patients with grade < 2 anemia had longer median OS than those with grade ≥ 2 anemia (35.00 vs. 25.10 months, P < 0.001). Multivariate analyses identified baseline anemia as a factor predicting a poor prognosis in terms of OS in patients with EGFR mutations. CONCLUSIONS Baseline anemia is a significant factor predicting a poor prognosis in terms of OS in patients with NSCLC and EGFR mutations treated with targeted therapy. A higher grade of baseline anemia may also be related to shorter OS. And a higher risk of EGFR-mutated patients who had received targeted therapy could also be observed.
Collapse
|
99884
|
Tang Q, Liang B, Zhang L, Li X, Li H, Jing W, Jiang Y, Zhou F, Zhang J, Meng Y, Yang X, Yang H, Huang G, Zhao J. Enhanced CHOLESTEROL biosynthesis promotes breast cancer metastasis via modulating CCDC25 expression and neutrophil extracellular traps formation. Sci Rep 2022; 12:17350. [PMID: 36253427 PMCID: PMC9576744 DOI: 10.1038/s41598-022-22410-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/14/2022] [Indexed: 01/10/2023] Open
Abstract
Neutrophil extracellular traps (NETs) has been demonstrated to regulate the metastasis of breast cancer. In this study, we showed that de novo cholesterol biosynthesis induced by ASPP2 depletion in mouse breast cancer cell 4T1 and human breast cancer cell MDA-MB-231 promoted NETs formation in vitro, as well as in lung metastases in mice intravenously injected with ASPP2-deficient 4T1 cells. Simvastatin and berberine (BBR), cholesterol synthesis inhibitors, efficiently blocked ASPP2-depletion induced NETs formation. Cholesterol biosynthesis greatly enhanced Coiled-coil domain containing protein 25 (CCDC25) expression on cancer cells as well as in lung metastases. CCDC25 expression was co-localized with caveolin-1, a lipid raft molecule, and was damped by inhibitor of lipid rafts formation. Our data suggest that cholesterol biosynthesis promotes CCDC25 expression in a lipid raft-dependent manner. Clinically, the expression of CCDC25 was positively correlated with the expression of 3-hydroxy-3-methylglutaryl-CoAreductase (HMRCG), and citrullinated histone H3 (H3cit), in tissues from breast cancer patients. High expression of CCDC25 and HMGCR was related with worse prognosis in breast cancer patients. In conclusion, our study explores a novel mechanism for de novo cholesterol biosynthesis in the regulation of CCDC25 expression, NETs formation and breast cancer metastasis. Targeting cholesterol biosynthesis may be promising therapeutic strategies to treat breast cancer metastasis.
Collapse
Affiliation(s)
- Qiqi Tang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.39436.3b0000 0001 2323 5732Shanghai University of Traditional Medicine, Shanghai, 201203 China
| | - Beibei Liang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Lisha Zhang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.39436.3b0000 0001 2323 5732Shanghai University of Traditional Medicine, Shanghai, 201203 China
| | - Xuhui Li
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Hengyu Li
- grid.411525.60000 0004 0369 1599Changhai Hospital, Navy Military Medical University, Shanghai, 200438 China
| | - Wei Jing
- grid.411525.60000 0004 0369 1599Changhai Hospital, Navy Military Medical University, Shanghai, 200438 China
| | - Yingjie Jiang
- grid.411525.60000 0004 0369 1599Changhai Hospital, Navy Military Medical University, Shanghai, 200438 China
| | - Felix Zhou
- grid.4991.50000 0004 1936 8948Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ UK
| | - Jian Zhang
- grid.8547.e0000 0001 0125 2443Phase I Clinical Trial Center, Shanghai Cancer Center, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Yanchun Meng
- grid.8547.e0000 0001 0125 2443Phase I Clinical Trial Center, Shanghai Cancer Center, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Xinhua Yang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.39436.3b0000 0001 2323 5732Shanghai University of Traditional Medicine, Shanghai, 201203 China
| | - Hao Yang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Gang Huang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Jian Zhao
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China ,grid.39436.3b0000 0001 2323 5732Shanghai University of Traditional Medicine, Shanghai, 201203 China
| |
Collapse
|
99885
|
Abstract
DNA damage by chemicals, radiation, or oxidative stress leads to a mutational spectrum, which is complex because it is determined in part by lesion structure, the DNA sequence context of the lesion, lesion repair kinetics, and the type of cells in which the lesion is replicated. Accumulation of mutations may give rise to genetic diseases such as cancer and therefore understanding the process underlying mutagenesis is of immense importance to preserve human health. Chemical or physical agents that cause cancer often leave their mutational fingerprints, which can be used to back-calculate the molecular events that led to disease. To make a clear link between DNA lesion structure and the mutations a given lesion induces, the field of single-lesion mutagenesis was developed. In the last three decades this area of research has seen much growth in several directions, which we attempt to describe in this Perspective.
Collapse
Affiliation(s)
- Ashis K Basu
- Department of Chemistry, The University of Connecticut Storrs, Storrs, Connecticut 06269, United States
| | - John M Essigmann
- Departments of Chemistry, Biological Engineering and Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
99886
|
Majumder R, Ghosh S, Das A, Singh MK, Samanta S, Saha A, Saha RP. Prokaryotic ncRNAs: Master regulators of gene expression. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100136. [PMID: 36568271 PMCID: PMC9780080 DOI: 10.1016/j.crphar.2022.100136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/11/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022] Open
Abstract
ncRNA plays a very pivotal role in various biological activities ranging from gene regulation to controlling important developmental networks. It is imperative to note that this small molecule is not only present in all three domains of cellular life, but is an important modulator of gene regulation too in all these domains. In this review, we discussed various aspects of ncRNA biology, especially their role in bacteria. The last two decades of scientific research have proved that this molecule plays an important role in the modulation of various regulatory pathways in bacteria including the adaptive immune system and gene regulation. It is also very surprising to note that this small molecule is also employed in various processes related to the pathogenicity of virulent microorganisms.
Collapse
Affiliation(s)
- Rajib Majumder
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Sanmitra Ghosh
- Department of Biological Sciences, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Arpita Das
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Manoj Kumar Singh
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Saikat Samanta
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Abinit Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India,Corresponding authors.
| | - Rudra P. Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India,Corresponding authors.
| |
Collapse
|
99887
|
Zhao J, Jiang H, Zou G, Lin Q, Wang Q, Liu J, Ma L. CNNArginineMe: A CNN structure for training models for predicting arginine methylation sites based on the One-Hot encoding of peptide sequence. Front Genet 2022; 13:1036862. [PMID: 36324513 PMCID: PMC9618650 DOI: 10.3389/fgene.2022.1036862] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/04/2022] [Indexed: 11/30/2022] Open
Abstract
Protein arginine methylation (PRme), as one post-translational modification, plays a critical role in numerous cellular processes and regulates critical cellular functions. Though several in silico models for predicting PRme sites have been reported, new models may be required to develop due to the significant increase of identified PRme sites. In this study, we constructed multiple machine-learning and deep-learning models. The deep-learning model CNN combined with the One-Hot coding showed the best performance, dubbed CNNArginineMe. CNNArginineMe performed best in AUC scoring metrics in comparisons with several reported predictors. Additionally, we employed CNNArginineMe to predict arginine methylation proteome and performed functional analysis. The arginine methylated proteome is significantly enriched in the amyotrophic lateral sclerosis (ALS) pathway. CNNArginineMe is freely available at https://github.com/guoyangzou/CNNArginineMe.
Collapse
Affiliation(s)
- Jiaojiao Zhao
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Haoqiang Jiang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Guoyang Zou
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qian Lin
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Qiang Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Leina Ma
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao University, Qingdao, China
- *Correspondence: Leina Ma,
| |
Collapse
|
99888
|
Andrés CMC, Pérez de la Lastra JM, Juan CA, Plou FJ, Pérez-Lebeña E. The Role of Reactive Species on Innate Immunity. Vaccines (Basel) 2022; 10:vaccines10101735. [PMID: 36298601 PMCID: PMC9609844 DOI: 10.3390/vaccines10101735] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/06/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
This review examines the role of reactive species RS (of oxygen ROS, nitrogen RNS and halogen RHS) on innate immunity. The importance of these species in innate immunity was first recognized in phagocytes that underwent a “respiratory burst” after activation. The anion superoxide •O2− and hydrogen peroxide H2O2 are detrimental to the microbial population. NADPH oxidase NOx, as an •O2− producer is essential for microbial destruction, and patients lacking this functional oxidase are more susceptible to microbial infections. Reactive nitrogen species RNS (the most important are nitric oxide radical -•NO, peroxynitrite ONOO— and its derivatives), are also harmful to microorganisms, including bacteria, viruses, and parasites. Hypochlorous acid HOCl and hypothiocyanous acid HOSCN synthesized through the enzyme myeloperoxidase MPO, which catalyzes the reaction between H2O2 and Cl− or SCN−, are important inorganic bactericidal molecules, effective against a wide range of microbes. This review also discusses the role of antimicrobial peptides AMPs and their induction of ROS. In summary, reactive species RS are the heart of the innate immune system, and they are necessary for microbial lysis in infections that can affect mammals throughout their lives.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez 3, 38206 La Laguna, Spain
- Correspondence:
| | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén 7, 47011 Valladolid, Spain
| | - Francisco J. Plou
- Institute of Catalysis and Petrochemistry, CSIC-Spanish Research Council, 28049 Madrid, Spain
| | | |
Collapse
|
99889
|
Yu C, Jiang L, Yang D, Dong X, Yu R, Yu H. Anlotinib Hydrochloride and PD-1 Blockade as a Salvage Second-Line Treatment in Patients with Progress of Local Advanced Non-Small Cell Lung Cancer in Half a Year After Standard Treatment. Onco Targets Ther 2022; 15:1221-1228. [PMID: 36262804 PMCID: PMC9575589 DOI: 10.2147/ott.s380615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/01/2022] [Indexed: 11/10/2022] Open
Abstract
Purpose As for local advanced non-small cell lung cancer (NSCLC), synchronous radiotherapy and chemotherapy is the standard treatment mode. But for patients with progress in half a year, which means the second-line chemotherapy effect is not ideal for them. We observed the efficacy and safety of anlotinib hydrochloride combined with PD-1 blockade as the second-line treatment for those patients in this trial. Patients and Methods From January 2018 to December 2019, 57 patients with the progress of local advanced NSCLC treated with anlotinib plus PD-1 blockade until disease progression or intolerance as a result of adverse events. Patients have been assessed using computed tomography prior to treatment and during follow-up every 2 months until disease progression or death. The primary endpoint was objective response rate (ORR). The secondary endpoints included overall survival (OS), progression-free survival (PFS) and safety. Survival curves were created using the Kaplan-Meier method. Results 57 patients were enrolled. The median age was 64 years, and 61.4% of the patients were men. The ORR was 50.9% with a median OS time of 14 months and the 1-year OS rates and PFS rates were 81.8% and 33.3%, respectively. The patients with squamous cell carcinoma, no brain or liver metastases had longer PFS than patients with liver metastasis. When the PFS was calculated from the time of second treatment, the median PFS was 9 months. Most adverse events (AEs) were grade 1-3, one drug-related death was noted. Conclusion The expected outcome of this study is that anlotinib combined with PD-1 blockade has tolerable toxicity and better ORR, OS than second-line chemotherapy. The results may indicate additional treatment options for patients with progress of local advance NSCLC in half a year after standard treatment.
Collapse
Affiliation(s)
- Chengqi Yu
- School of Basic Medical Science, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Leilei Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Dan Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Xin Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Rong Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Huiming Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China,Correspondence: Huiming Yu, Department of Radiation Oncology, Peking University Cancer Hospital & Institute, 52# Fucheng Road, Haidian District, Beijing, 100142, People’s Republic of China, Tel +86 13699249320, Fax +86 10-59300192, Email
| |
Collapse
|
99890
|
Croft W, Evans RPT, Pearce H, Elshafie M, Griffiths EA, Moss P. The single cell transcriptional landscape of esophageal adenocarcinoma and its modulation by neoadjuvant chemotherapy. Mol Cancer 2022; 21:200. [PMID: 36253784 PMCID: PMC9575245 DOI: 10.1186/s12943-022-01666-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 09/17/2022] [Indexed: 11/10/2022] Open
Abstract
Immune checkpoint blockade has recently proven effective in subsets of patients with esophageal adenocarcinoma (EAC) but little is known regarding the EAC immune microenvironment. We determined the single cell transcriptional profile of EAC in 8 patients who were treatment-naive (n = 4) or had received neoadjuvant chemotherapy (n = 4). Analysis of 52,387 cells revealed 10 major cell subsets of tumor, immune and stromal cells. Prior to chemotherapy tumors were heavy infiltrated by T regulatory cells and exhausted effector T cells whilst plasmacytoid dendritic cells were markedly expanded. Two dominant cancer-associated fibroblast populations were also observed whilst endothelial populations were suppressed. Pathological remission following chemotherapy associated with broad reversal of immune abnormalities together with fibroblast transition and an increase in endothelial cells whilst a chemoresistant epithelial stem cell population correlated with poor response. These findings reveal features that underlie and limit the response to current immunotherapy and identify a range of novel opportunities for targeted therapy.
Collapse
Affiliation(s)
- Wayne Croft
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Centre for Computational Biology, University of Birmingham, Birmingham, UK
| | - Richard P T Evans
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- University Hospitals Foundation Trust, Edgbaston, Birmingham, UK
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Mona Elshafie
- University Hospitals Foundation Trust, Edgbaston, Birmingham, UK
| | - Ewen A Griffiths
- University Hospitals Foundation Trust, Edgbaston, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
- University Hospitals Foundation Trust, Edgbaston, Birmingham, UK.
| |
Collapse
|
99891
|
Waudby CA, Alvarez-Teijeiro S, Josue Ruiz E, Suppinger S, Pinotsis N, Brown PR, Behrens A, Christodoulou J, Mylona A. An intrinsic temporal order of c-JUN N-terminal phosphorylation regulates its activity by orchestrating co-factor recruitment. Nat Commun 2022; 13:6133. [PMID: 36253406 PMCID: PMC9576782 DOI: 10.1038/s41467-022-33866-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 10/05/2022] [Indexed: 12/24/2022] Open
Abstract
Protein phosphorylation is a major regulatory mechanism of cellular signalling. The c-JUN proto-oncoprotein is phosphorylated at four residues within its transactivation domain (TAD) by the JNK family kinases, but the functional significance of c-JUN multisite phosphorylation has remained elusive. Here we show that c-JUN phosphorylation by JNK exhibits defined temporal kinetics, with serine63 and serine73 being phosphorylated more rapidly than threonine91 and threonine93. We identify the positioning of the phosphorylation sites relative to the kinase docking motif, and their primary sequence, as the main factors controlling phosphorylation kinetics. Functional analysis reveals three c-JUN phosphorylation states: unphosphorylated c-JUN recruits the MBD3 repressor, serine63/73 doubly-phosphorylated c-JUN binds to the TCF4 co-activator, whereas the fully phosphorylated form disfavours TCF4 binding attenuating JNK signalling. Thus, c-JUN phosphorylation encodes multiple functional states that drive a complex signalling response from a single JNK input.
Collapse
Affiliation(s)
- Christopher A Waudby
- Institute of Structural and Molecular Biology, University College London, London, UK
- School of Pharmacy, University College London, London, UK
| | - Saul Alvarez-Teijeiro
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, UK
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - E Josue Ruiz
- Cancer Stem Cell Laboratory, Institute of Cancer Research, London, UK
| | - Simon Suppinger
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, UK
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | - Nikos Pinotsis
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, UK
| | - Paul R Brown
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College, London, UK
| | - Axel Behrens
- Cancer Stem Cell Laboratory, Institute of Cancer Research, London, UK
- Division of Cancer, Department of Surgery and Cancer, Imperial College, London, UK
- CR-UK Convergence Science Centre, Imperial College, London, SW7 2BU, UK
| | - John Christodoulou
- Institute of Structural and Molecular Biology, University College London, London, UK.
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, UK.
| | - Anastasia Mylona
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, UK.
- Division of Cancer, Department of Surgery and Cancer, Imperial College, London, UK.
| |
Collapse
|
99892
|
IGFBP3 inhibits tumor growth and invasion of lung cancer cells and is associated with improved survival in lung cancer patients. Transl Oncol 2022; 27:101566. [PMID: 36257207 PMCID: PMC9583099 DOI: 10.1016/j.tranon.2022.101566] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/27/2022] [Accepted: 10/10/2022] [Indexed: 11/15/2022] Open
Abstract
The insulin-like growth factor (IGF)-pathway is involved in tumor cell proliferation, metastasis, and survival. We aimed to find out what effects IGF binding protein 3 (IGFBP3) exerted on H1299 lung cancer (LC) cells in terms of tumor growth and invasion and whether IGFBP3 was associated with clinical and pathological parameters in a prospective cohort of LC patients. H1299 cells were transfected with an IGFBP3-expressing vector. Its influence on apoptosis induction via flow cytometry annexin V FITC assay, cell proliferation in 2D and 3D cell culture, and invasion were examined. Expression of several matrix metalloproteinases (MMPs) and inhibitors (TIMP-1) were also investigated in IGFBP3-transfected LC cells. Further, data on LC patients (n = 131), tumor characteristics, and survival were prospectively collected and correlated with IGFBP3 plasma levels. IGFBP3 did not influence apoptosis induction and 2D cell proliferation. However, both spheroid growth (3D proliferation) and invasion of IGFBP3-transfected cells planted in an extracellular matrix-based gel were significantly inhibited. IGFBP3 inhibited MMP-1 release, and the total MMP activity. In LC patients, higher IGFBP3 plasma levels correlated with both lower clinical tumor stage, grading, Ki-67 staining, and the absence of necrosis (P < 0.05, respectively). Increased IGFBP3 plasma levels were associated with improved overall survival (hazard ratio 0.37, P = 0.01). In conclusion, overexpressed IGFBP3 in a LC cell line inhibited tumor growth and invasion. Translating from bench to bedside, investigation of clinicopathological parameters confirmed these experimental results showing that higher IGFBP3 plasma levels were associated with less aggressive tumor growth, reduced tumor spread, and improved survival of LC patients.
Collapse
|
99893
|
Shi Y, Gao Q, Liu Z, Shen G, Sun X, Di X. Identification of Immune and Hypoxia Risk Classifier to Estimate Immune Microenvironment and Prognosis in Cervical Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6906380. [PMID: 36304989 PMCID: PMC9593224 DOI: 10.1155/2022/6906380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/04/2022] [Accepted: 10/01/2022] [Indexed: 11/07/2023]
Abstract
Purpose Cervical cancer (CC) is one of the most common gynecologic neoplasms. Hypoxia is an essential trigger for activating immunosuppressive activity and initiating malignant tumors. However, the determination of the role of immunity and hypoxia on the clinical outcome of CC patients remains unclear. Methods The CC independent cohort were collected from TCGA database. Consensus cluster analysis was employed to determine a molecular subtype based on immune and hypoxia gene sets. Cox relevant analyses were utilized to set up a risk classifier for prognosis assessment. The underlying pathways of classifier genes were detected by GSEA. Moreover, we conducted CIBERSORT algorithm to mirror the immune status of CC samples. Results We observed two cluster related to immune and hypoxia status and found the significant difference in outcome of patients between the two clusters. A total of 251 candidate genes were extracted from the two clusters and enrolled into Cox relevant analyses. Then, seven hub genes (CCL20, CXCL2, ITGA5, PLOD2, PTGS2, TGFBI, and VEGFA) were selected to create an immune and hypoxia-based risk classifier (IHBRC). The IHBRC can precisely distinguish patient risk and estimate clinical outcomes. In addition, IHBRC was closely bound up with tumor associated pathways such as hypoxia, P53 signaling and TGF β signaling. IHBRC was also tightly associated with numerous types of immunocytes. Conclusion This academic research revealed that IHBRC can be served as predictor for prognosis assessment and cancer treatment estimation in CC.
Collapse
Affiliation(s)
- Yujing Shi
- Department of Oncology, Jurong People's Hospital, Huayang Town, Jurong City, China
| | - Qing Gao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zeyuan Liu
- Department of Radiation Oncology, Nanjing Jiangning Hospital and the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Gefenqiang Shen
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinchen Sun
- Department of Oncology, Jurong People's Hospital, Huayang Town, Jurong City, China
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoke Di
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
99894
|
Zhang Q, Ma L, Zhou H, Zhou Y, Liu S, Li Q. A prognostic signature of cuproptosis and TCA-related genes for hepatocellular carcinoma. Front Oncol 2022; 12:1040736. [PMID: 36324575 PMCID: PMC9619237 DOI: 10.3389/fonc.2022.1040736] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/03/2022] [Indexed: 11/26/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most common malignant tumor of the liver. Cuproptosis is a newly defined form of cell death. Copper ion induces cell death by binding to the tricarboxylic acid cycle (TCA). The effect of cuproptosis-related and TCA-related genes on the clinical prognosis of HCC is still unclear. In this study, we explores the genetic changes of cuproptosis-related genes that affect the TCA process and their potential therapeutic value in HCC patients. Methods The cuproptosis and TCA-related genes were obtained from cuproptosis-related articles and the molecular signatures database. The prognosis signatures of eight related genes were constructed using the last absolute shrinkage and selection operator (LASSO), and Receiver Operating Characteristic (ROC) curves were used to evaluate the signature. In addition, we analyzed downstream functional enrichment and immune infiltration to explore cuproptosis-inducing drugs and immunotherapeutic responses. All these analyses were validated using multiple datasets of the International Cancer Genome Consortium (ICGC). Results TCA and copper malnutrition-related genes (CDKN2A, IDH1, OGDHL, IDH3G, IDH3B, GLS, DLAT, LIPT1) were finally included. According to the risk score, they were divided into high-risk and low-risk groups. Survival analysis showed that the overall survival (OS) of the high-risk group was significantly lower than that of the low-risk group. We established a risk prognostic feature to predict the OS of patients with HCC. Based on this feature and the clinical stage, we constructed a nomogram. Functional enrichment analysis revealed pathways related to organelle division and the cell cycle. Different risk scores had different immune abundances in immune cells (including macrophages and regulatory T-cells) and immune pathways (including antigen-presenting cells co-stimulation). Moreover, the drug sensitivity of eleschomol and PD-L1 in the high-risk group was better than that in the low-risk group. The status of TP53 somatic mutation was also closely related to the risk score. Conclusion In this study, we established a new prediction signature of eight genes related to cuproptosis and the TCA process, which can effectively predict the prognosis of HCC patients.
Collapse
|
99895
|
Moser JC, Salvador E, Deniz K, Swanson K, Tuszynski J, Carlson KW, Karanam NK, Patel CB, Story M, Lou E, Hagemann C. The Mechanisms of Action of Tumor Treating Fields. Cancer Res 2022; 82:3650-3658. [PMID: 35839284 PMCID: PMC9574373 DOI: 10.1158/0008-5472.can-22-0887] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/24/2022] [Accepted: 07/13/2022] [Indexed: 01/07/2023]
Abstract
Tumor treating fields (TTFields), a new modality of cancer treatment, are electric fields transmitted transdermally to tumors. The FDA has approved TTFields for the treatment of glioblastoma multiforme and mesothelioma, and they are currently under study in many other cancer types. While antimitotic effects were the first recognized biological anticancer activity of TTFields, data have shown that tumor treating fields achieve their anticancer effects through multiple mechanisms of action. TTFields therefore have the ability to be useful for many cancer types in combination with many different treatment modalities. Here, we review the current understanding of TTFields and their mechanisms of action.
Collapse
Affiliation(s)
- Justin C. Moser
- HonorHealth Research and Innovation Institute, Scottsdale, Arizona.,Department of Medicine, University of Arizona College of Medicine- Phoenix, Phoenix, Arizona.,Corresponding Author: Justin Moser, HonorHealth Research and Innovation Institute, 10510 N 92nd Street Ste 200, Scottsdale, AZ 85258. Phone: 480-323-4638, E-mail:
| | - Ellaine Salvador
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| | - Karina Deniz
- Department of Medicine, Division of Hematology Oncology and Transplant, University of Minnesota, Minneapolis, Minnesota
| | - Kenneth Swanson
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Jack Tuszynski
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Kristen W. Carlson
- Department of Neurosurgery, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts
| | - Narasimha Kumar Karanam
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chirag B. Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston Texas.,Neuroscience and Cancer Biology Graduate Programs, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences
| | - Michael Story
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Emil Lou
- Department of Medicine, Division of Hematology Oncology and Transplant, University of Minnesota, Minneapolis, Minnesota
| | - Carsten Hagemann
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| |
Collapse
|
99896
|
Li S, Wang L, Wang Y, Zhang C, Hong Z, Han Z. The synthetic lethality of targeting cell cycle checkpoints and PARPs in cancer treatment. J Hematol Oncol 2022; 15:147. [PMID: 36253861 PMCID: PMC9578258 DOI: 10.1186/s13045-022-01360-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Continuous cell division is a hallmark of cancer, and the underlying mechanism is tumor genomics instability. Cell cycle checkpoints are critical for enabling an orderly cell cycle and maintaining genome stability during cell division. Based on their distinct functions in cell cycle control, cell cycle checkpoints are classified into two groups: DNA damage checkpoints and DNA replication stress checkpoints. The DNA damage checkpoints (ATM-CHK2-p53) primarily monitor genetic errors and arrest cell cycle progression to facilitate DNA repair. Unfortunately, genes involved in DNA damage checkpoints are frequently mutated in human malignancies. In contrast, genes associated with DNA replication stress checkpoints (ATR-CHK1-WEE1) are rarely mutated in tumors, and cancer cells are highly dependent on these genes to prevent replication catastrophe and secure genome integrity. At present, poly (ADP-ribose) polymerase inhibitors (PARPi) operate through “synthetic lethality” mechanism with mutant DNA repair pathways genes in cancer cells. However, an increasing number of patients are acquiring PARP inhibitor resistance after prolonged treatment. Recent work suggests that a combination therapy of targeting cell cycle checkpoints and PARPs act synergistically to increase the number of DNA errors, compromise the DNA repair machinery, and disrupt the cell cycle, thereby increasing the death rate of cancer cells with DNA repair deficiency or PARP inhibitor resistance. We highlight a combinational strategy involving PARP inhibitors and inhibition of two major cell cycle checkpoint pathways, ATM-CHK2-TP53 and ATR-CHK1-WEE1. The biological functions, resistance mechanisms against PARP inhibitors, advances in preclinical research, and clinical trials are also reviewed.
Collapse
Affiliation(s)
- Shuangying Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Liangliang Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yuanyuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Changyi Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhenya Hong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Zhiqiang Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
99897
|
Li H, Gu J, Tian Y, Li S, Zhang H, Dai Z, Wang Z, Zhang N, Peng R. A prognostic signature consisting of metabolism-related genes and SLC17A4 serves as a potential biomarker of immunotherapeutic prediction in prostate cancer. Front Immunol 2022; 13:982628. [PMID: 36325340 PMCID: PMC9620963 DOI: 10.3389/fimmu.2022.982628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Prostate cancer (PCa), a prevalent malignant cancer in males worldwide, screening for patients might benefit more from immuno-/chemo-therapy remained inadequate and challenging due to the heterogeneity of PCa patients. Thus, the study aimed to explore the metabolic (Meta) characteristics and develop a metabolism-based signature to predict the prognosis and immuno-/chemo-therapy response for PCa patients. METHODS Differentially expressed genes were screened among 2577 metabolism-associated genes. Univariate Cox analysis and random forest algorithms was used for features screening. Multivariate Cox regression analysis was conducted to construct a prognostic Meta-model based on all combinations of metabolism-related features. Then the correlation between MetaScore and tumor was deeply explored from prognostic, genomic variant, functional and immunological perspectives, and chemo-/immuno-therapy response. Multiple algorithms were applied to estimate the immunotherapeutic responses of two MeteScore groups. Further in vitro functional experiments were performed using PCa cells to validate the association between the expression of hub gene SLC17A4 which is one of the model component genes and tumor progression. GDSC database was employed to determine the sensitivity of chemotherapy drugs. RESULTS Two metabolism-related clusters presented different features in overall survival (OS). A metabolic model was developed weighted by the estimated regression coefficients in the multivariate Cox regression analysis (0.5154*GAS2 + 0.395*SLC17A4 - 0.1211*NTM + 0.2939*GC). This Meta-scoring system highlights the relationship between the metabolic profiles and genomic alterations, gene pathways, functional annotation, and tumor microenvironment including stromal, immune cells, and immune checkpoint in PCa. Low MetaScore is correlated with increased mutation burden and microsatellite instability, indicating a superior response to immunotherapy. Several medications that might improve patients` prognosis in the MetaScore group were identified. Additionally, our cellular experiments suggested knock-down of SLC17A4 contributes to inhibiting invasion, colony formation, and proliferation in PCa cells in vitro. CONCLUSIONS Our study supports the metabolism-based four-gene signature as a novel and robust model for predicting prognosis, and chemo-/immuno-therapy response in PCa patients. The potential mechanisms for metabolism-associated genes in PCa oncogenesis and progression were further determined.
Collapse
Affiliation(s)
- He Li
- The Animal Laboratory Center, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jie Gu
- Department of Geriatric Urology, Xiangya International Medical Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Yuqiu Tian
- Department of Infectious Disease, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Shuyu Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Nan Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- One‑Third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Hei Longjiang, China
| | - Renjun Peng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
99898
|
Sun X, Zhang Y, Li H, Zhou Y, Shi S, Chen Z, He X, Zhang H, Li F, Yin J, Mou M, Wang Y, Qiu Y, Zhu F. DRESIS: the first comprehensive landscape of drug resistance information. Nucleic Acids Res 2022; 51:D1263-D1275. [PMID: 36243960 PMCID: PMC9825618 DOI: 10.1093/nar/gkac812] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/22/2022] [Accepted: 10/11/2022] [Indexed: 01/30/2023] Open
Abstract
Widespread drug resistance has become the key issue in global healthcare. Extensive efforts have been made to reveal not only diverse diseases experiencing drug resistance, but also the six distinct types of molecular mechanisms underlying this resistance. A database that describes a comprehensive list of diseases with drug resistance (not just cancers/infections) and all types of resistance mechanisms is now urgently needed. However, no such database has been available to date. In this study, a comprehensive database describing drug resistance information named 'DRESIS' was therefore developed. It was introduced to (i) systematically provide, for the first time, all existing types of molecular mechanisms underlying drug resistance, (ii) extensively cover the widest range of diseases among all existing databases and (iii) explicitly describe the clinically/experimentally verified resistance data for the largest number of drugs. Since drug resistance has become an ever-increasing clinical issue, DRESIS is expected to have great implications for future new drug discovery and clinical treatment optimization. It is now publicly accessible without any login requirement at: https://idrblab.org/dresis/.
Collapse
Affiliation(s)
| | | | | | | | - Shuiyang Shi
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xin He
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China,Zhejiang University–University of Edinburgh Institute, Zhejiang University, Haining 314499, China
| | - Hanyu Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiayi Yin
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yunzhu Wang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yunqing Qiu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- To whom correspondence should be addressed.
| |
Collapse
|
99899
|
Sun T, Li Y, Yang Y, Liu B, Cao Y, Yang W. Enhanced radiation-induced immunogenic cell death activates chimeric antigen receptor T cells by targeting CD39 against glioblastoma. Cell Death Dis 2022; 13:875. [PMID: 36245000 PMCID: PMC9573869 DOI: 10.1038/s41419-022-05319-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 01/23/2023]
Abstract
Chimeric antigen receptor (CAR)-T cells directed to solid tumors have been less effective, due in part to the low or lost expression of specific tumor antigens. Herein, we developed a different strategy to enhance CAR-T cell persistence and efficacy by producing a multispecific CAR-T or vaccine based on immunogenic cell death (ICD). We demonstrated that ionizing radiation activates STAT1-IRF1-CD39 axis to upregulate CD39 expression to form an immunosuppressive tumor microenvironment (TME) to enhance radioresistance. CD39 blockade accumulates extracellular ATP, which activates NLRP3 inflammasome in dendritic cells via P2X7 receptor, thereby promoting radiation-induced ICD. Multispecific CAR-T cells in vitro prepared by elevated ICD suppress the growth of xenografts in nude mice. Radiation and CD39 inhibition-induced ICD of glioma stem cells as a vaccine enhance CAR-T expansion in peripheral blood, multifunctionality in the TME, and antitumor effect in a glioma model. The multispecificity of CAR-T cells, targeting CAR and tumor antigens, vastly enhances the function of conventional CAR-T cells, stimulates a native immune response, and overcomes obstacles of specific antigen loss or low expression of target cells in antitumor therapy.
Collapse
Affiliation(s)
- Ting Sun
- grid.429222.d0000 0004 1798 0228Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006 China
| | - Yanyan Li
- grid.429222.d0000 0004 1798 0228Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006 China
| | - Ying Yang
- grid.263761.70000 0001 0198 0694State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu China
| | - Bin Liu
- grid.429222.d0000 0004 1798 0228Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006 China
| | - Yufei Cao
- grid.429222.d0000 0004 1798 0228Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006 China
| | - Wei Yang
- grid.263761.70000 0001 0198 0694State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu China
| |
Collapse
|
99900
|
Mendes MPR, Paiva MJN, Costa-Amaral IC, Carvalho LVB, Figueiredo VO, Gonçalves ES, Larentis AL, André LC. Metabolomic Study of Urine from Workers Exposed to Low Concentrations of Benzene by UHPLC-ESI-QToF-MS Reveals Potential Biomarkers Associated with Oxidative Stress and Genotoxicity. Metabolites 2022; 12:metabo12100978. [PMID: 36295880 PMCID: PMC9611274 DOI: 10.3390/metabo12100978] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/23/2022] Open
Abstract
Benzene is a human carcinogen whose exposure to concentrations below 1 ppm (3.19 mg·m-3) is associated with myelotoxic effects. The determination of biomarkers such as trans-trans muconic acid (AttM) and S-phenylmercapturic acid (SPMA) show exposure without reflecting the toxic effects of benzene. For this reason, in this study, the urinary metabolome of individuals exposed to low concentrations of benzene was investigated, with the aim of understanding the biological response to exposure to this xenobiotic and identifying metabolites correlated with the toxic effects induced by it. Ultra-efficient liquid chromatography coupled to a quadrupole-time-of-flight mass spectrometer (UHPLC-ESI-Q-ToF-MS) was used to identify metabolites in the urine of environmentally (n = 28) and occupationally exposed (n = 32) to benzene (mean of 22.1 μg·m-3 and 31.8 μg·m-3, respectively). Non-targeted metabolomics analysis by PLS-DA revealed nine urinary metabolites discriminating between groups and statistically correlated with oxidative damage (MDA, thiol) and genetic material (chromosomal aberrations) induced by the hydrocarbon. The analysis of metabolic pathways revealed important alterations in lipid metabolism. These results point to the involvement of alterations in lipid metabolism in the mechanisms of cytotoxic and genotoxic action of benzene. Furthermore, this study proves the potential of metabolomics to provide relevant information to understand the biological response to exposure to xenobiotics and identify early effect biomarkers.
Collapse
Affiliation(s)
- Michele P. R. Mendes
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil
| | - Maria José N. Paiva
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil
| | - Isabele C. Costa-Amaral
- Center for the Study of Occupational Health and Human Ecology (CESTEH), Sergio Arouca National School of Public Health (ENSP), Oswaldo Cruz Foundation (Fiocruz), Rua Leopoldo Bulhões 1480, Manguinhos, Rio de Janeiro 21041-210, RJ, Brazil
| | - Leandro V. B. Carvalho
- Center for the Study of Occupational Health and Human Ecology (CESTEH), Sergio Arouca National School of Public Health (ENSP), Oswaldo Cruz Foundation (Fiocruz), Rua Leopoldo Bulhões 1480, Manguinhos, Rio de Janeiro 21041-210, RJ, Brazil
| | - Victor O. Figueiredo
- Center for the Study of Occupational Health and Human Ecology (CESTEH), Sergio Arouca National School of Public Health (ENSP), Oswaldo Cruz Foundation (Fiocruz), Rua Leopoldo Bulhões 1480, Manguinhos, Rio de Janeiro 21041-210, RJ, Brazil
| | - Eline S. Gonçalves
- Center for the Study of Occupational Health and Human Ecology (CESTEH), Sergio Arouca National School of Public Health (ENSP), Oswaldo Cruz Foundation (Fiocruz), Rua Leopoldo Bulhões 1480, Manguinhos, Rio de Janeiro 21041-210, RJ, Brazil
| | - Ariane L. Larentis
- Center for the Study of Occupational Health and Human Ecology (CESTEH), Sergio Arouca National School of Public Health (ENSP), Oswaldo Cruz Foundation (Fiocruz), Rua Leopoldo Bulhões 1480, Manguinhos, Rio de Janeiro 21041-210, RJ, Brazil
| | - Leiliane C. André
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil
- Correspondence: ; Tel.: +55-31-9238-3636
| |
Collapse
|