1
|
Dai Y, Li J, Yamamoto K, Goyama S, Loza M, Park SJ, Nakai K. Integrative analysis of cancer multimodality data identifying COPS5 as a novel biomarker of diffuse large B-cell lymphoma. Front Genet 2024; 15:1407765. [PMID: 38974382 PMCID: PMC11224480 DOI: 10.3389/fgene.2024.1407765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Preventing, diagnosing, and treating diseases requires accurate clinical biomarkers, which remains challenging. Recently, advanced computational approaches have accelerated the discovery of promising biomarkers from high-dimensional multimodal data. Although machine-learning methods have greatly contributed to the research fields, handling data sparseness, which is not unusual in research settings, is still an issue as it leads to limited interpretability and performance in the presence of missing information. Here, we propose a novel pipeline integrating joint non-negative matrix factorization (JNMF), identifying key features within sparse high-dimensional heterogeneous data, and a biological pathway analysis, interpreting the functionality of features by detecting activated signaling pathways. By applying our pipeline to large-scale public cancer datasets, we identified sets of genomic features relevant to specific cancer types as common pattern modules (CPMs) of JNMF. We further detected COPS5 as a potential upstream regulator of pathways associated with diffuse large B-cell lymphoma (DLBCL). COPS5 exhibited co-overexpression with MYC, TP53, and BCL2, known DLBCL marker genes, and its high expression was correlated with a lower survival probability of DLBCL patients. Using the CRISPR-Cas9 system, we confirmed the tumor growth effect of COPS5, which suggests it as a novel prognostic biomarker for DLBCL. Our results highlight that integrating multiple high-dimensional data and effectively decomposing them to interpretable dimensions unravels hidden biological importance, which enhances the discovery of clinical biomarkers.
Collapse
Affiliation(s)
- Yutong Dai
- Department of Computational Biology and Medical Science, The University of Tokyo, Kashiwa, Japan
| | - Jingmei Li
- Department of Computational Biology and Medical Science, The University of Tokyo, Kashiwa, Japan
| | - Keita Yamamoto
- Department of Computational Biology and Medical Science, The University of Tokyo, Kashiwa, Japan
| | - Susumu Goyama
- Department of Computational Biology and Medical Science, The University of Tokyo, Kashiwa, Japan
| | - Martin Loza
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sung-Joon Park
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Science, The University of Tokyo, Kashiwa, Japan
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Füzesi-Levi MG, Ben-Nissan G, Listov D, Fridmann Sirkis Y, Hayouka Z, Fleishman S, Sharon M. The C-terminal tail of CSNAP attenuates the CSN complex. Life Sci Alliance 2023; 6:e202201634. [PMID: 37460146 PMCID: PMC10355216 DOI: 10.26508/lsa.202201634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023] Open
Abstract
Protein degradation is one of the essential mechanisms that enables reshaping of the proteome landscape in response to various stimuli. The largest E3 ubiquitin ligase family that targets proteins to degradation by catalyzing ubiquitination is the cullin-RING ligases (CRLs). Many of the proteins that are regulated by CRLs are central to tumorigenesis and tumor progression, and dysregulation of the CRL family is frequently associated with cancer. The CRL family comprises ∼300 complexes, all of which are regulated by the COP9 signalosome complex (CSN). Therefore, CSN is considered an attractive target for therapeutic intervention. Research efforts for targeted CSN inhibition have been directed towards inhibition of the complex enzymatic subunit, CSN5. Here, we have taken a fresh approach focusing on CSNAP, the smallest CSN subunit. Our results show that the C-terminal region of CSNAP is tightly packed within the CSN complex, in a groove formed by CSN3 and CSN8. We show that a 16 amino acid C-terminal peptide, derived from this CSN-interacting region, can displace the endogenous CSNAP subunit from the complex. This, in turn, leads to a CSNAP null phenotype that attenuates CSN activity and consequently CRLs function. Overall, our findings emphasize the potential of a CSNAP-based peptide for CSN inhibition as a new therapeutic avenue.
Collapse
Affiliation(s)
- Maria G Füzesi-Levi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Gili Ben-Nissan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Dina Listov
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Sarel Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
3
|
Repurposing Drugs in Small Animal Oncology. Animals (Basel) 2022; 13:ani13010139. [PMID: 36611747 PMCID: PMC9817697 DOI: 10.3390/ani13010139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Repurposing drugs in oncology consists of using off-label drugs that are licensed for various non-oncological medical conditions to treat cancer. Repurposing drugs has the advantage of using drugs that are already commercialized, with known mechanisms of action, proven safety profiles, and known toxicology, pharmacokinetics and pharmacodynamics, and posology. These drugs are usually cheaper than new anti-cancer drugs and thus more affordable, even in low-income countries. The interest in repurposed anti-cancer drugs has led to numerous in vivo and in vitro studies, with some promising results. Some randomized clinical trials have also been performed in humans, with certain drugs showing some degree of clinical efficacy, but the true clinical benefit for most of these drugs remains unknown. Repurposing drugs in veterinary oncology is a very new concept and only a few studies have been published so far. In this review, we summarize both the benefits and challenges of using repurposed anti-cancer drugs; we report and discuss the most relevant studies that have been previously published in small animal oncology, and we suggest potential drugs that could be clinically investigated for anti-cancer treatment in dogs and cats.
Collapse
|
4
|
Rapier-Sharman N, Clancy J, Pickett BE. Joint Secondary Transcriptomic Analysis of Non-Hodgkin's B-Cell Lymphomas Predicts Reliance on Pathways Associated with the Extracellular Matrix and Robust Diagnostic Biomarkers. JOURNAL OF BIOINFORMATICS AND SYSTEMS BIOLOGY : OPEN ACCESS 2022; 5:119-135. [PMID: 36873459 PMCID: PMC9980876 DOI: 10.26502/jbsb.5107040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Approximately 450,000 cases of Non-Hodgkin's lymphoma are annually diagnosed worldwide, resulting in ~240,000 deaths. An augmented understanding of the common mechanisms of pathology among larger numbers of B-cell Non-Hodgkin's Lymphoma (BCNHL) patients is sorely needed. We consequently performed a large joint secondary transcriptomic analysis of the available BCNHL RNA-sequencing projects from GEO, consisting of 322 relevant samples across ten distinct public studies, to find common underlying mechanisms and biomarkers across multiple BCNHL subtypes and patient subpopulations; limitations may include lack of diversity in certain ethnicities and age groups and limited clinical subtype diversity due to sample availability. We found ~10,400 significant differentially expressed genes (FDR-adjusted p-value < 0.05) and 33 significantly modulated pathways (Bonferroni-adjusted p-value < 0.05) when comparing BCNHL samples to non-diseased B-cell samples. Our findings included a significant class of proteoglycans not previously associated with lymphomas as well as significant modulation of genes that code for extracellular matrix-associated proteins. Our drug repurposing analysis predicted new candidates for repurposed drugs including ocriplasmin and collagenase. We also used a machine learning approach to identify robust BCNHL biomarkers that include YES1, FERMT2, and FAM98B, which have not previously been associated with BCNHL in the literature, but together provide ~99.9% combined specificity and sensitivity for differentiating lymphoma cells from healthy B-cells based on measurement of transcript expression levels in B-cells. This analysis supports past findings and validates existing knowledge while providing novel insights into the inner workings and mechanisms of transformed B-cell lymphomas that could give rise to improved diagnostics and/or therapeutics.
Collapse
Affiliation(s)
- Naomi Rapier-Sharman
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - Jeffrey Clancy
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - Brett E Pickett
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
5
|
Qi B, Boscenco S, Ramamurthy J, Trakadis YJ. Transcriptomics and machine learning to advance schizophrenia genetics: A case-control study using post-mortem brain data. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 214:106590. [PMID: 34954633 DOI: 10.1016/j.cmpb.2021.106590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 08/31/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND AND OBJECTIVE Alterations of the expression of a variety of genes have been reported in patients with schizophrenia (SCZ). Moreover, machine learning (ML) analysis of gene expression microarray data has shown promising preliminary results in the study of SCZ. Our objective was to evaluate the performance of ML in classifying SCZ cases and controls based on gene expression microarray data from the dorsolateral prefrontal cortex. METHODS We apply a state-of-the-art ML algorithm (XGBoost) to train and evaluate a classification model using 201 SCZ cases and 278 controls. We utilized 10-fold cross-validation for model selection, and a held-out testing set to evaluate the model. The performance metric utilizes to evaluate classification performance was the area under the receiver-operator characteristics curve (AUC). RESULTS We report an average AUC on 10-fold cross-validation of 0.76 and an AUC of 0.76 on testing data, not used during training. Analysis of the rolling balanced classification accuracy from high to low prediction confidence levels showed that the most certain subset of predictions ranged between 80-90%. The ML model utilized 182 gene expression probes. Further improvement to classification performance was observed when applying an automated ML strategy on the 182 features, which achieved an AUC of 0.79 on the same testing data. We found literature evidence linking all of the top ten ML ranked genes to SCZ. Furthermore, we leveraged information from the full set of microarray gene expressions available via univariate differential gene expression analysis. We then prioritized differentially expressed gene sets using the piano gene set analysis package. We augmented the ranking of the prioritized gene sets with genes from the complex multivariate ML model using hypergeometric tests to identify more robust gene sets. We identified two significant Gene Ontology molecular function gene sets: "oxidoreductase activity, acting on the CH-NH2 group of donors" and "integrin binding." Lastly, we present candidate treatments for SCZ based on findings from our study CONCLUSIONS: Overall, we observed above-chance performance from ML classification of SCZ cases and controls based on brain gene expression microarray data, and found that ML analysis of gene expressions could further our understanding of the pathophysiology of SCZ and help identify novel treatments.
Collapse
Affiliation(s)
- Bill Qi
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Sonia Boscenco
- Faculty of Science, McGill University, Montreal, QC, Canada
| | | | - Yannis J Trakadis
- Department of Human Genetics, McGill University, Montreal, QC, Canada; Department of Medical Genetics, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
6
|
Lei H, Wang J, Hu J, Zhu Q, Wu Y. Deubiquitinases in hematological malignancies. Biomark Res 2021; 9:66. [PMID: 34454635 PMCID: PMC8401176 DOI: 10.1186/s40364-021-00320-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
Deubiquitinases (DUBs) are enzymes that control the stability, interactions or localization of most cellular proteins by removing their ubiquitin modification. In recent years, some DUBs, such as USP7, USP9X and USP10, have been identified as promising therapeutic targets in hematological malignancies. Importantly, some potent inhibitors targeting the oncogenic DUBs have been developed, showing promising inhibitory efficacy in preclinical models, and some have even undergone clinical trials. Different DUBs perform distinct function in diverse hematological malignancies, such as oncogenic, tumor suppressor or context-dependent effects. Therefore, exploring the biological roles of DUBs and their downstream effectors will provide new insights and therapeutic targets for the occurrence and development of hematological malignancies. We summarize the DUBs involved in different categories of hematological malignancies including leukemia, multiple myeloma and lymphoma. We also present the recent development of DUB inhibitors and their applications in hematological malignancies. Together, we demonstrate DUBs as potential therapeutic drug targets in hematological malignancies.
Collapse
Affiliation(s)
- Hu Lei
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jiaqi Wang
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiacheng Hu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Zhu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingli Wu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
7
|
Jab1/Cops5: a promising target for cancer diagnosis and therapy. Int J Clin Oncol 2021; 26:1159-1169. [PMID: 34019195 DOI: 10.1007/s10147-021-01933-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 05/08/2021] [Indexed: 01/21/2023]
Abstract
C-Jun activation domain-binding protein1 (Jab1), the fifth component of the constitutive photomorphogenic-9 signalosome (COPS5/Csn5) complex, functions in several cellular processes to affect different signaling pathways. Dysregulation of Jab1/Csn5 both restrains tumor suppressors and activates oncogenes to contribute oncogenesis. Jab1 overexpressed in various tumors and played an essential part in cancer initiation, progression and prognosis, which has spurred strong research interest in developing inhibitors for cancer therapy. Here, we summarize the multiple signaling pathways and functions of Jab1/Csn5 in tumorigenesis. By querying the Oncomine database, a cancer microarray database and web-based data-mining platform aimed at facilitating discovery from genome-wide expression analyses, we investigated statistically the differential expression of Jab1/Csn5 between different cancer samples and the corresponding normal tissue samples, cancer samples with different histological types, different cancer types, and different clinical outcomes. These statistical data confirmed the significant role of Jab1/Csn5 in carcinogenesis, indicating Jab1/Csn5 as a biomarker and a therapeutic target in different cancers.
Collapse
|
8
|
Wang Z, Wang Y, Yang H, Guo J, Wang Z. Doxycycline Induces Apoptosis of Brucella Suis S2 Strain-Infected HMC3 Microglial Cells by Activating Calreticulin-Dependent JNK/p53 Signaling Pathway. Front Cell Infect Microbiol 2021; 11:640847. [PMID: 33996626 PMCID: PMC8113685 DOI: 10.3389/fcimb.2021.640847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 04/13/2021] [Indexed: 01/18/2023] Open
Abstract
Neurobrucellosis is a chronic complication of human brucellosis that is caused by the presence of Brucella spp in the central nervous system (CNS) and the inflammation play a key role on the pathogenesis. Doxycycline (Dox) is a widely used antibiotic that induces apoptosis of bacteria-infected cells. However, the mechanisms of Brucella inhibition of microglial apoptosis and Dox induction of apoptosis are still poorly understood. In this study, we found that Brucella suis S2 strain (B. suis S2) increased calreticulin (CALR) protein levels and inhbited HMC3 cell apoptosis. Hence, we constructed two HMC3 cell line variants, one with stable overexpression (HMC3-CALR) and one with low expression of CALR (HMC3-sh-CALR). CALR was found to decrease levels of p-JNK and p-p53 proteins, as well as suppress apoptosis in HMC3 cells. These findings suggest that CALR suppresses apoptosis by inhibiting the JNK/p53 signaling pathway. Next, we treated HMC3, HMC3-CALR and HMC3-sh-CALR cell lines with B. suis S2 or Dox. Our results demonstrate that B. suis S2 restrains the JNK/p53 signaling pathway to inhibit HMC3 cell apoptosis via increasing CALR protein expression, while Dox plays the opposite role. Finally, we treated B. suis S2-infected HMC3 cells with Dox. Our results confirm that Dox induces JNK/p53-dependent apoptosis in B. suis S2-infected HMC3 cells through inhibition of CALR protein expression. Taken together, these results reveal that CALR and the JNK/p53 signaling pathway may serve as novel therapeutic targets for treatment of neurobrucellosis.
Collapse
Affiliation(s)
- Zhao Wang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yanbai Wang
- Cerebrospinal Fluid Laboratory, The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Huan Yang
- Emergency Department, The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jiayu Guo
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Zhenhai Wang
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, China.,Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, The General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
9
|
Blondelle J, Biju A, Lange S. The Role of Cullin-RING Ligases in Striated Muscle Development, Function, and Disease. Int J Mol Sci 2020; 21:E7936. [PMID: 33114658 PMCID: PMC7672578 DOI: 10.3390/ijms21217936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
The well-orchestrated turnover of proteins in cross-striated muscles is one of the fundamental processes required for muscle cell function and survival. Dysfunction of the intricate protein degradation machinery is often associated with development of cardiac and skeletal muscle myopathies. Most muscle proteins are degraded by the ubiquitin-proteasome system (UPS). The UPS involves a number of enzymes, including E3-ligases, which tightly control which protein substrates are marked for degradation by the proteasome. Recent data reveal that E3-ligases of the cullin family play more diverse and crucial roles in cross striated muscles than previously anticipated. This review highlights some of the findings on the multifaceted functions of cullin-RING E3-ligases, their substrate adapters, muscle protein substrates, and regulatory proteins, such as the Cop9 signalosome, for the development of cross striated muscles, and their roles in the etiology of myopathies.
Collapse
Affiliation(s)
- Jordan Blondelle
- Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Andrea Biju
- Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Stephan Lange
- Department of Medicine, University of California, La Jolla, CA 92093, USA
- Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| |
Collapse
|
10
|
Garg M. Epithelial Plasticity, Autophagy and Metastasis: Potential Modifiers of the Crosstalk to Overcome Therapeutic Resistance. Stem Cell Rev Rep 2020; 16:503-510. [PMID: 32125607 DOI: 10.1007/s12015-019-09945-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) initiates malignant transformation of cancer cells and is responsible for the generation of heterogenic subsets of cancer stem cells (CSCs). Signals in the form of environmental cues and paracrine factors within tumor microenvironment (TME) niche, support the possibility of generation of pool of CSCs with two distinct functional transition states. Cyclic CSCs with predominant epithelial phenotype, self-renew and differentiate into mature cancer cells. Subsets of autophagic/ non-cyclic CSCs with predominant mesenchymal phenotype have capacity to invade, metastasize, resist to apoptosis, escape immunosurveillance, survive chemotherapies and are majorly responsible for cancer mortality. Differences in phenotypic plasticity may form the basis of differential impact of therapeutic outcomes on heterogeneous subpopulations of CSCs. Activation of autophagy is responsible for the recycling of damaged organelles and protein aggregates, regulates EMT, confers the survival advantage to neoplastic cells to anti-cancer therapies, significantly affects the invasive potential of cancer cells and supports their metastatic dissemination in a tissue and tumor stage dependent manner. Therapy resistance is the primary obstacle in the complete ablation of tumor cells. Combinational treatments based on targeting autophagic CSCs and inhibiting EMT regulators may represent potential anticancer strategies for the prevention of cancer invasion, metastatic spread and disease relapse.
Collapse
Affiliation(s)
- Minal Garg
- Department of Biochemistry, University of Lucknow, Lucknow, 226007, India.
| |
Collapse
|
11
|
Melenotte C, Mezouar S, Mège JL, Gorvel JP, Kroemer G, Raoult D. Bacterial infection and non-Hodgkin's lymphoma. Crit Rev Microbiol 2020; 46:270-287. [PMID: 32412856 DOI: 10.1080/1040841x.2020.1760786] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
One quarter of all cancers are linked to infectious diseases. The link between viral infection and cancer has been widely studied, but few reports have focused on the carcinogenic role of bacterial infection. Nonetheless, Helicobacter pylori, Chlamydia psittaci, Coxiella burnetii, Borrelia burgdorferi and Campylobacter jejuni are bacteria that can be associated with non-Hodgkin's lymphoma (NHL), the most common haematologic malignancy. Here, we review the evidence in favour of a link between these bacterial infections and NHL. Sero-epidemiological observation makes it possible to identify a link between H. pylori, C. burnetii, B. burgdorferi infection and NHL. Helicobacter pylori, Chlamydia psittaci, Coxiella burnetii, Borrelia burgdorferi and Campylobacter jejuni could be identified in NHL tissue samples at the site of chronic inflammation, where B and T lymphocytes are attracted to participate in follicle formation. Lymphoma remissions have been observed under antimicrobial therapies supporting the carcinogenic contribution of bacteria. If the theory of causality is characterized by the lack of universal criteria for establishing a causal link between two diseases, infection and lymphoma, epidemiological, clinical, and histological evidences reported here, should lead clinicians to pay attention to these infectious agents, to detect early lymphoma transformation.
Collapse
Affiliation(s)
- Cléa Melenotte
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| | - Jean-Louis Mège
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| | | | - Guido Kroemer
- Cell Biology and Metabolomics platforms, Villejuif, France.,INSERM, Paris, France.,Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Didier Raoult
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
12
|
Mitochondria as target to inhibit proliferation and induce apoptosis of cancer cells: the effects of doxycycline and gemcitabine. Sci Rep 2020; 10:4363. [PMID: 32152409 PMCID: PMC7063048 DOI: 10.1038/s41598-020-61381-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/25/2020] [Indexed: 01/09/2023] Open
Abstract
Doxycycline has anti-tumour effects in a range of tumour systems. The aims of this study were to define the role mitochondria play in this process and examine the potential of doxycycline in combination with gemcitabine. We studied the adenocarcinoma cell line A549, its mitochondrial DNA-less derivative A549 ρ° and cultured fibroblasts. Treatment with doxycycline for 5 days resulted in a decrease of mitochondrial-encoded proteins, respiration and membrane potential, and an increase of reactive oxygen species in A549 cells and fibroblasts, but fibroblasts were less affected. Doxycycline slowed proliferation of A549 cells by 35%. Cellular ATP levels did not change. Doxycycline alone had no effect on apoptosis; however, in combination with gemcitabine given during the last 2 days of treatment, doxycycline increased caspase 9 and 3/7 activities, resulting in a further decrease of surviving A549 cells by 59% and of fibroblasts by 24% compared to gemcitabine treatment alone. A549 ρ° cells were not affected by doxycycline. Key effects of doxycycline observed in A549 cells, such as the decrease of mitochondrial-encoded proteins and surviving cells were also seen in the cancer cell lines COLO357 and HT29. Our results suggest that doxycycline suppresses cancer cell proliferation and primes cells for apoptosis by gemcitabine.
Collapse
|
13
|
Armando RG, Gómez DLM, Gomez DE. New drugs are not enough‑drug repositioning in oncology: An update. Int J Oncol 2020; 56:651-684. [PMID: 32124955 PMCID: PMC7010222 DOI: 10.3892/ijo.2020.4966] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022] Open
Abstract
Drug repositioning refers to the concept of discovering novel clinical benefits of drugs that are already known for use treating other diseases. The advantages of this are that several important drug characteristics are already established (including efficacy, pharmacokinetics, pharmacodynamics and toxicity), making the process of research for a putative drug quicker and less costly. Drug repositioning in oncology has received extensive focus. The present review summarizes the most prominent examples of drug repositioning for the treatment of cancer, taking into consideration their primary use, proposed anticancer mechanisms and current development status.
Collapse
Affiliation(s)
- Romina Gabriela Armando
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Diego Luis Mengual Gómez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Daniel Eduardo Gomez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| |
Collapse
|
14
|
Pandian J, Panneerpandian P, Devanandan HJ, Sekar BT, Balakrishnan K, Selvarasu K, Muthupandi K, Ganesan K. Identification of the targeted therapeutic potential of doxycycline for a subset of gastric cancer patients. Ann N Y Acad Sci 2020; 1467:94-111. [PMID: 31944316 DOI: 10.1111/nyas.14288] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 12/26/2022]
Abstract
The identification of new drugs for the targeted therapy of gastric cancer remains an important need. The RAS/RAF/MEK/ERK/ELK1 signaling cascade is activated in many cancers, including gastric cancer. To identify the targetable inhibitors of the ERK/MAPK pathway, we performed a repurposing screening of a panel of antimicrobial agents in gastric cancer cells using an ERK/MAPK-driven firefly luciferase reporter assay. Multiple antibiotics were identified to inhibit ERK-mediated transcriptional activity. Among them, doxycycline showed high inhibition of ERK/MAPK-regulated transcriptional activity and the levels of ERK proteins. Doxycycline was further identified to inhibit the proliferation and the colony- and spheroid-forming potential of gastric cancer cells. By in vitro signaling pathway and genome-wide expression profiling analyses, doxycycline was identified to inhibit signaling pathways and transcriptional activities regulated by ER, Myc, E2F1, Wnt, SMAD2/3/4, Notch, and OCT4. Doxycycline was also found to activate p53-, ATF6-, NRF1/2-, and MTF1-mediated transcription and inhibit the transcription of histones, proteasomal genes, fibroblast growth factor, and other oncogenic factors. These observations show the multitargeting and targeted therapeutic features of doxycycline for a subset of gastric tumors.
Collapse
Affiliation(s)
- Jaishree Pandian
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Ponmathi Panneerpandian
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Helen Jemimah Devanandan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Balaji T Sekar
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Karthik Balakrishnan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Karthikeyan Selvarasu
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Karthikeyan Muthupandi
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Kumaresan Ganesan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| |
Collapse
|
15
|
Escanilla C, Guavita Falla PM, Cevallos C, Ávalos Jobet N, Bobadilla Bruneau F. Primary cutaneous CD4-positive small/medium T-cell lymphoproliferative disorder: The first-reported Latin-American case with response to doxycycline. Clin Case Rep 2019; 7:2405-2409. [PMID: 31893069 PMCID: PMC6935602 DOI: 10.1002/ccr3.2454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/12/2019] [Accepted: 08/22/2019] [Indexed: 12/18/2022] Open
Abstract
Primary cutaneous CD4+ small/medium T-cell lymphoproliferative disorder is a provisional entity according to the last WHO-EORTC classification. The treatment of choice has not yet been defined. Local therapies have been used with variable response. Doxycycline as a main treatment option is a potential low-cost and effective alternative for this disorder.
Collapse
Affiliation(s)
| | | | - Carolina Cevallos
- Departamento de DermatologíaUniversidad de Santiago de ChileSantiagoChile
| | | | | |
Collapse
|
16
|
El Sayed H, Shalaby S, Abdel-Halim MRE, Aboelfadl DM, Samir N. Efficacy of doxycycline in the treatment of early stages of mycosis fungoides: a randomized controlled trial. J DERMATOL TREAT 2019; 32:424-431. [PMID: 31526286 DOI: 10.1080/09546634.2019.1667474] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Mycosis fungoides is the most common type of primary cutaneous T cell lymphomas. Doxycycline promoted apoptosis in different human malignant cell lines and in vivo models. OBJECTIVES To test for the therapeutic efficacy of doxycycline in comparison to PUVA in early stages of classic MF and its effect on T cell apoptosis. METHODS Thirty-six patients were randomized into either: doxycycline 200 mg daily (n = 18) or PUVA (3 weekly sessions) (n = 18) for 12 weeks. The primary outcome (therapeutic efficacy) was defined in terms of objective response rate (ORR) which was measured according to changes in the modified severity weighted assessment tool (mSWAT). RESULTS Doxycycline achieved significantly less ORR (partial response) in comparison to PUVA (11.1%, 50%, respectively, p = .016). The percent reduction in mSWAT, CAILS, histopathology score and CD3 expression was significantly less in the doxycycline group (p = .001, p = .001, p ˂ .001, and p = .004, respectively). Within the doxycycline group, changes in mSWAT and CAILS showed no correlation with changes in the CD3 or Bcl-2 expression. Gastric upset was significantly more encountered in the doxycycline group (p = .001). CONCLUSION Doxycycline is not suitable as a sole agent in the treatment of early stages of classic MF, acting mainly by anti-inflammatory rather apoptotic function. REGISTER NUMBER NCT03454945 (www.clinicaltrials.gov).
Collapse
Affiliation(s)
- H El Sayed
- Dermatology Department, Faculty of Medicine, Kasr Al Aini University Hospital, Cairo University, Giza, Egypt
| | - S Shalaby
- Dermatology Department, Faculty of Medicine, Kasr Al Aini University Hospital, Cairo University, Giza, Egypt
| | - M R E Abdel-Halim
- Dermatology Department, Faculty of Medicine, Kasr Al Aini University Hospital, Cairo University, Giza, Egypt
| | - D M Aboelfadl
- Pathology Department, National Research Institute, Cairo, Egypt
| | - N Samir
- Dermatology Department, Faculty of Medicine, Kasr Al Aini University Hospital, Cairo University, Giza, Egypt
| |
Collapse
|
17
|
CSNAP, the smallest CSN subunit, modulates proteostasis through cullin-RING ubiquitin ligases. Cell Death Differ 2019; 27:984-998. [PMID: 31367012 DOI: 10.1038/s41418-019-0392-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 11/09/2022] Open
Abstract
The cullin-RING ubiquitin E3 ligase (CRL) family consists of ~250 complexes that catalyze ubiquitylation of proteins to achieve cellular regulation. All CRLs are inhibited by the COP9 signalosome complex (CSN) through both enzymatic (deneddylation) and nonenzymatic (steric) mechanisms. The relative contribution of these two mechanisms is unclear. Here, we decouple the mechanisms using CSNAP, the recently discovered ninth subunit of the CSN. We find that CSNAP reduces the affinity of CSN toward CRL complexes. Removing CSNAP does not affect deneddylation, but leads to global effects on the CRL, causing altered reproductive capacity, suppressed DNA damage response, and delayed cell cycle progression. Thus, although CSNAP is only 2% of the CSN mass, it plays a critical role in the steric regulation of CRLs by the CSN.
Collapse
|
18
|
Ubiquitination of MBNL1 Is Required for Its Cytoplasmic Localization and Function in Promoting Neurite Outgrowth. Cell Rep 2019; 22:2294-2306. [PMID: 29490267 DOI: 10.1016/j.celrep.2018.02.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/19/2017] [Accepted: 02/06/2018] [Indexed: 12/20/2022] Open
Abstract
The Muscleblind-like protein family (MBNL) plays an important role in regulating the transition between differentiation and pluripotency and in the pathogenesis of myotonic dystrophy type 1 (DM1), a CTG expansion disorder. How different MBNL isoforms contribute to the differentiation and are affected in DM1 has not been investigated. Here, we show that the MBNL1 cytoplasmic, but not nuclear, isoform promotes neurite morphogenesis and reverses the morphological defects caused by expanded CUG RNA. Cytoplasmic MBNL1 is polyubiquitinated by lysine 63 (K63). Reduced cytoplasmic MBNL1 in the DM1 mouse brain is consistent with the reduced extent of K63 ubiquitination. Expanded CUG RNA induced the deubiqutination of cytoplasmic MBNL1, which resulted in nuclear translocation and morphological impairment that could be ameliorated by inhibiting K63-linked polyubiquitin chain degradation. Our results suggest that K63-linked ubiquitination of MBNL1 is required for its cytoplasmic localization and that deubiquitination of cytoplasmic MBNL1 is pathogenic in the DM1 brain.
Collapse
|
19
|
CSN5 inhibition triggers inflammatory signaling and Rho/ROCK-dependent loss of endothelial integrity. Sci Rep 2019; 9:8131. [PMID: 31148579 PMCID: PMC6544660 DOI: 10.1038/s41598-019-44595-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 05/16/2019] [Indexed: 12/23/2022] Open
Abstract
RhoGTPases regulate cytoskeletal dynamics, migration and cell-cell adhesion in endothelial cells. Besides regulation at the level of guanine nucleotide binding, they also undergo post-translational modifications, for example ubiquitination. RhoGTPases are ubiquitinated by Cullin RING ligases which are in turn regulated by neddylation. Previously we showed that inhibition of Cullin RING ligase activity by the neddylation inhibitor MLN4924 is detrimental for endothelial barrier function, due to accumulation of RhoB and the consequent induction of contractility. Here we analyzed the effect of pharmacological activation of Cullin RING ligases on endothelial barrier integrity in vitro and in vivo. CSN5i-3 induced endothelial barrier disruption and increased macromolecule leakage in vitro and in vivo. Mechanistically, CSN5i-3 strongly induced the expression and activation of RhoB and to lesser extent of RhoA in endothelial cells, which enhanced cell contraction. Elevated expression of RhoGTPases was a consequence of activation of the NF-κB pathway. In line with this notion, CSN5i-3 treatment decreased IκBα expression and increased NF-κB-mediated ICAM-1 expression and consequent adhesion of neutrophils to endothelial cells. This study shows that sustained neddylation of Cullin RING-ligases leads to activation the NF-κB pathway in endothelial cells, elevated expression of RhoGTPases, Rho/ROCK-dependent activation of MLC and disruption of the endothelial barrier.
Collapse
|
20
|
Guo Z, Wang Y, Zhao Y, Shu Y, Liu Z, Zhou H, Wang H, Zhang W. The pivotal oncogenic role of Jab1/CSN5 and its therapeutic implications in human cancer. Gene 2018; 687:219-227. [PMID: 30468907 DOI: 10.1016/j.gene.2018.11.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/01/2018] [Accepted: 11/19/2018] [Indexed: 01/28/2023]
Abstract
Jab1/CSN5 is a conserved multifunctional protein involved in ubiquitin-mediated protein degradation. Deregulation of Jab1/CSN5 can exert dramatic effects on diverse cellular functions, including DNA repair, cell cycle control, apoptosis, angiogenesis, and signal transduction, all of which are critical for tumor development. Although increasing evidence has demonstrated that Jab1/CSN5 was overexpressed in a variety of human cancers and usually correlated with poor prognosis, little was known about the underlying regulatory principles that coordinated its function. In this review, we highlight recent advances of the oncogenic role of Jab1/CSN5 and its potential as a therapeutic target for anticancer intervention.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Youhong Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Yu Zhao
- Key Laboratory of Translational Radiation Oncology, Hunan Province, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, PR China
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Hui Wang
- Key Laboratory of Translational Radiation Oncology, Hunan Province, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, PR China.
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| |
Collapse
|
21
|
Protasoni M, Kroon AM, Taanman JW. Mitochondria as oncotarget: a comparison between the tetracycline analogs doxycycline and COL-3. Oncotarget 2018; 9:33818-33831. [PMID: 30333912 PMCID: PMC6173462 DOI: 10.18632/oncotarget.26107] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/24/2018] [Indexed: 01/23/2023] Open
Abstract
Tetracyclines have anticancer properties in addition to their well-known antibacterial properties. It has been proposed that tetracyclines slow metastasis and angiogenesis through inhibition of matrix metalloproteinases. However, we believe that the anticancer effect of tetracyclines is due to their inhibition of mitochondrial protein synthesis, resulting in a decrease of the mitochondrial energy generating capacity. Several groups have developed analogs that are void of antibacterial action. An example is COL-3, which is currently tested for its anticancer effects in clinical trials. We have undertaken a comparative study of the tetracycline analogs COL-3 and doxycycline, which has an antibacterial function, to further investigate the role of the mitochondrial energy generating capacity in the anticancer mechanism and, thereby, evaluate the usefulness of mitochondria as an oncotarget. Our experiments with cultures of the human A549, COLO357 and HT29 cancer cells and fibroblasts indicated that COL-3 is significantly more cytotoxic than doxycycline. Mitochondrial translation assays demonstrated that COL-3 has retained its inhibitory effect on mitochondrial protein synthesis. Both drugs caused a severe decrease in the levels of mitochondrially encoded cytochrome-c oxidase subunits and cytochrome-c oxidase activity. In addition, COL-3 produced a marked drop in the level of nuclear-encoded succinate dehydrogenase subunit A and citrate synthase activity, indicating that COL-3 has multiple inhibitory effects. Contrary to COL-3, the anticancer action of doxycycline appears to be based specifically on inhibition of mitochondrial protein synthesis, which is thought to affect rapidly proliferating cancer cells more than healthy tissue. Doxycycline is likely to cause less side effects that COL-3.
Collapse
Affiliation(s)
- Margherita Protasoni
- Department of Clinical and Movement Neurosciences, Institute of Neurology, University College London, London, NW3 2PF, UK
| | - Albert M Kroon
- Department of Clinical and Movement Neurosciences, Institute of Neurology, University College London, London, NW3 2PF, UK
| | - Jan-Willem Taanman
- Department of Clinical and Movement Neurosciences, Institute of Neurology, University College London, London, NW3 2PF, UK
| |
Collapse
|
22
|
Abstract
Inhibitors of mTOR, including clinically available rapalogs such as rapamycin (Sirolimus) and Everolimus, are gerosuppressants, which suppress cellular senescence. Rapamycin slows aging and extends life span in a variety of species from worm to mammals. Rapalogs can prevent age-related diseases, including cancer, atherosclerosis, obesity, neurodegeneration and retinopathy and potentially rejuvenate stem cells, immunity and metabolism. Here, I further suggest how rapamycin can be combined with metformin, inhibitors of angiotensin II signaling (Losartan, Lisinopril), statins (simvastatin, atorvastatin), propranolol, aspirin and a PDE5 inhibitor. Rational combinations of these drugs with physical exercise and an anti-aging diet (Koschei formula) can maximize their anti-aging effects and decrease side effects.
Collapse
|
23
|
Doxycycline is an NF-κB inhibitor that induces apoptotic cell death in malignant T-cells. Oncotarget 2018; 7:75954-75967. [PMID: 27732942 PMCID: PMC5342790 DOI: 10.18632/oncotarget.12488] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 09/24/2016] [Indexed: 12/24/2022] Open
Abstract
Cutaneous T-cell Lymphoma (CTCL) is a rare non-Hodgkin's lymphoma that can affect the skin, blood, and lymph nodes, and can metastasize at late stages. Novel therapies that target all affected disease compartments and provide longer lasting responses while being safe are needed. One potential therapeutic target is NF-κB, a regulator of immune responses and an important participant in carcinogenesis and cancer progression. As a transcription factor, NF-κB targets genes that promote cell proliferation and survival. Constitutive or aberrant activation of NF-κB is encountered in many types of cancer, including CTCL.Recently, while analyzing gene-expression profiles of a variety of small molecule compounds that target NF-κB, we discovered the tetracycline family of antibiotics, including doxycycline, to be potent inhibitors of the NF-κB pathway. Doxycycline is well-tolerated, safe, and inexpensive; and is commonly used as an antibiotic and anti-inflammatory for the treatment a multitude of medical conditions.In our current study, we show that doxycycline induces apoptosis in a dose dependent manner in multiple different cell lines from patients with the two most common subtypes of CTCL, Mycosis Fungoides (MF) and Sézary Syndrome (SS). Similar results were found using primary CD4+ T cells from a patient with SS. Doxycycline inhibits TNF induced NF-κB activation and reduces expression of NF-κB dependent anti-apoptotic proteins, such as BCL2α. Furthermore, we have identified that doxycycline induces apoptosis through reactive oxygen species.
Collapse
|
24
|
Hume KR, Sylvester SR, Borlle L, Balkman CE, McCleary-Wheeler AL, Pulvino M, Casulo C, Zhao J. Metabolic Abnormalities Detected in Phase II Evaluation of Doxycycline in Dogs with Multicentric B-Cell Lymphoma. Front Vet Sci 2018. [PMID: 29536017 PMCID: PMC5834767 DOI: 10.3389/fvets.2018.00025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Doxycycline has antiproliferative effects in human lymphoma cells and in murine xenografts. We hypothesized that doxycycline would decrease canine lymphoma cell viability and prospectively evaluated its clinical tolerability in client-owned dogs with spontaneous, nodal, multicentric, substage a, B-cell lymphoma, not previously treated with chemotherapy. Treatment duration ranged from 1 to 8 weeks (median and mean, 3 weeks). Dogs were treated with either 10 (n = 6) or 7.5 (n = 7) mg/kg by mouth twice daily. One dog had a stable disease for 6 weeks. No complete or partial tumor responses were observed. Five dogs developed grade 3 and/or 4 metabolic abnormalities suggestive of hepatopathy with elevations in bilirubin, ALT, ALP, and/or AST. To evaluate the absorption of oral doxycycline in our study population, serum concentrations in 10 treated dogs were determined using liquid chromatography tandem mass spectrometry. Serum levels were variable and ranged from 3.6 to 16.6 µg/ml (median, 7.6 µg/ml; mean, 8.8 µg/ml). To evaluate the effect of doxycycline on canine lymphoma cell viability in vitro, trypan blue exclusion assay was performed on canine B-cell lymphoma cell lines (17-71 and CLBL) and primary B-cell lymphoma cells from the nodal tissue of four dogs. A doxycycline concentration of 6 µg/ml decreased canine lymphoma cell viability by 80%, compared to matched, untreated, control cells (mixed model analysis, p < 0.0001; Wilcoxon signed rank test, p = 0.0313). Although the short-term administration of oral doxycycline is not associated with the remission of canine lymphoma, combination therapy may be worthwhile if future research determines that doxycycline can alter cell survival pathways in canine lymphoma cells. Due to the potential for metabolic abnormalities, close monitoring is recommended with the use of this drug in tumor-bearing dogs. Additional research is needed to assess the tolerability of chronic doxycycline therapy.
Collapse
Affiliation(s)
- Kelly R Hume
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Skylar R Sylvester
- College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Lucia Borlle
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Cheryl E Balkman
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Angela L McCleary-Wheeler
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Mary Pulvino
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
| | - Carla Casulo
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, United States
| | - Jiyong Zhao
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
25
|
Melenotte C, Raoult D. Pro-apoptotic effect of doxycycline and hydroxychloroquine on B-cell lymphoma induced by C. burnetii. Oncotarget 2017; 9:2726-2727. [PMID: 29416805 PMCID: PMC5788673 DOI: 10.18632/oncotarget.23397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 12/16/2017] [Indexed: 11/25/2022] Open
Affiliation(s)
- Cléa Melenotte
- Aix Marseille Université, CNRS, IRD, INSERM, AP-HM, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, Institut Hospitalier Universitaire Méditerranée-Infection, Marseille, France
| | - Didier Raoult
- Aix Marseille Université, CNRS, IRD, INSERM, AP-HM, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, Institut Hospitalier Universitaire Méditerranée-Infection, Marseille, France
| |
Collapse
|
26
|
Zhou Z, Fang Q, Ma D, Zhe N, Ren M, Cheng B, Li P, Liu P, Lin X, Tang S, Hu X, Liao Y, Zhang Y, Lu T, Wang J. Silencing heme oxygenase-1 increases the sensitivity of ABC-DLBCL cells to histone deacetylase inhibitor in vitro and in vivo. Oncotarget 2017; 8:78480-78495. [PMID: 29108243 PMCID: PMC5667976 DOI: 10.18632/oncotarget.19652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 05/23/2017] [Indexed: 01/04/2023] Open
Abstract
Heme oxygenase-1 (HO-1) can promote tumor growth and reinforce the resistance of diffuse large B-cell lymphoma (DLBCL) cells to chemotherapeutic drug vincristine. We herein found that HO-1 protein expression was higher in high-risk DLBCL patients than in low-risk ones. Silencing HO-1 gene expression resisted vorinostat-induced apoptosis and arrested cell cycle in the G0/G1 phase of LY-10 cells. Western blot, co-immunoprecipitation and chromatin immunoprecipitation assays confirmed that the possible mechanisms may be increased cleaved caspase-3 protein expression, decreased phospho-histone deacetylase 3 protein expression, and activated histone acetylation of P27Kip1 promoter. Moreover, silencing HO-1 gene expression enhanced vorinostat-induced tumor cell apoptosis, prolonged survival time and promoted P27Kip1 protein expression in a xenograft mouse model. In conclusion, HO-1 is a potential therapeutic target of DLBCL. The findings provide a valuable preclinical evidence for sensitizing DLBCL patients with poor prognosis to histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Zhen Zhou
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China.,Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China.,Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China.,Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Nana Zhe
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Mei Ren
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Bingqing Cheng
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Peifan Li
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Ping Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Xiaojing Lin
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Sishi Tang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Xiuying Hu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Yudan Liao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Yaming Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Tingting Lu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treatment Centre of Guizhou Province, Guiyang 550004, China.,Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| |
Collapse
|
27
|
Lleonart ME, Grodzicki R, Graifer DM, Lyakhovich A. Mitochondrial dysfunction and potential anticancer therapy. Med Res Rev 2017; 37:1275-1298. [DOI: 10.1002/med.21459] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 12/11/2022]
Affiliation(s)
| | - Robert Grodzicki
- Thomas Steitz Laboratory; Department of Molecular Biophysics & Biochemistry, Center for Structural Biology, Howard Hughes Medical Institute; Yale University; New Haven Connecticut
| | | | - Alex Lyakhovich
- Oncology Program; Vall D'Hebron Research Institute; Barcelona Spain
- Institute of Molecular Biology and Biophysics, Novosibirsk; Russia
- International Clinical Research Center and St. Anne's University Hospital Brno; Czech Republic
| |
Collapse
|
28
|
Axelsson AS, Tubbs E, Mecham B, Chacko S, Nenonen HA, Tang Y, Fahey JW, Derry JMJ, Wollheim CB, Wierup N, Haymond MW, Friend SH, Mulder H, Rosengren AH. Sulforaphane reduces hepatic glucose production and improves glucose control in patients with type 2 diabetes. Sci Transl Med 2017; 9:9/394/eaah4477. [DOI: 10.1126/scitranslmed.aah4477] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 02/23/2017] [Accepted: 05/05/2017] [Indexed: 12/13/2022]
|
29
|
Tang X, Wang X, Zhao YY, Curtis JM, Brindley DN. Doxycycline attenuates breast cancer related inflammation by decreasing plasma lysophosphatidate concentrations and inhibiting NF-κB activation. Mol Cancer 2017; 16:36. [PMID: 28178994 PMCID: PMC5299726 DOI: 10.1186/s12943-017-0607-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/30/2017] [Indexed: 12/11/2022] Open
Abstract
Background We previously discovered that tetracyclines increase the expression of lipid phosphate phosphatases at the surface of cells. These enzymes degrade circulating lysophosphatidate and therefore doxycycline increases the turnover of plasma lysophosphatidate and decreases its concentration. Extracellular lysophosphatidate signals through six G protein-coupled receptors and it is a potent promoter of tumor growth, metastasis and chemo-resistance. These effects depend partly on the stimulation of inflammation that lysophosphatidate produces. Methods In this work, we used a syngeneic orthotopic mouse model of breast cancer to determine the impact of doxycycline on circulating lysophosphatidate concentrations and tumor growth. Cytokine/chemokine concentrations in tumor tissue and plasma were measured by multiplexing laser bead technology. Leukocyte infiltration in tumors was analyzed by immunohistochemistry. The expression of IL-6 in breast cancer cell lines was determined by RT-PCR. Cell growth was measured in Matrigel™ 3D culture. The effects of doxycycline on NF-κB-dependent signaling were analyzed by Western blotting. Results Doxycycline decreased plasma lysophosphatidate concentrations, delayed tumor growth and decreased the concentrations of several cytokines/chemokines (IL-1β, IL-6, IL-9, CCL2, CCL11, CXCL1, CXCL2, CXCL9, G-CSF, LIF, VEGF) in the tumor. These results were compatible with the effects of doxycycline in decreasing the numbers of F4/80+ macrophages and CD31+ blood vessel endothelial cells in the tumor. Doxycycline also decreased the lysophosphatidate-induced growth of breast cancer cells in three-dimensional culture. Lysophosphatidate-induced Ki-67 expression was inhibited by doxycycline. NF-κB activity in HEK293 cells transiently expressing a NF-κB-luciferase reporter vectors was also inhibited by doxycycline. Treatment of breast cancer cells with doxycycline also decreased the translocation of NF-κB to the nucleus and the mRNA levels for IL-6 in the presence or absence of lysophosphatidate. Conclusion These results contribute a new dimension for understanding the anti-inflammatory effects of tetracyclines, which make them potential candidates for adjuvant therapy of cancers and other inflammatory diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0607-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoyun Tang
- Department of Biochemistry, Signal Transduction Research Group, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Xianyan Wang
- Department of Biochemistry, Signal Transduction Research Group, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Yuan Y Zhao
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 410 Agriculture/Forestry Centre, 3-60D South Academic Building, Edmonton, AB, T6G 2P5, Canada
| | - Jonathan M Curtis
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 410 Agriculture/Forestry Centre, 3-60D South Academic Building, Edmonton, AB, T6G 2P5, Canada
| | - David N Brindley
- Department of Biochemistry, Signal Transduction Research Group, University of Alberta, Edmonton, AB, T6G 2S2, Canada. .,Department of Biochemistry, 357 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
30
|
Li P, Xie L, Gu Y, Li J, Xie J. Roles of Multifunctional COP9 Signalosome Complex in Cell Fate and Implications for Drug Discovery. J Cell Physiol 2017; 232:1246-1253. [PMID: 27869306 DOI: 10.1002/jcp.25696] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/18/2016] [Indexed: 01/24/2023]
Abstract
The eight subunits containing COP9 signalosome (CSN) complex, is highly conserved among eukaryotes. CSN, identified as a negative regulator of photomorphogenesis, has also been demonstrated to be important in proteolysis, cellular signal transduction and cell cycle regulation in various eukaryotic organisms. This review mainly summarizes the roles of CSN in cell cycle regulation, signal transduction and apoptosis, and its potential as diagnostic biomarkers, drug targets for cancer and infectious diseases. J. Cell. Physiol. 232: 1246-1253, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ping Li
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Longxiang Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Yinzhong Gu
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Jiang Li
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| |
Collapse
|
31
|
Zhang L, Xu L, Zhang F, Vlashi E. Doxycycline inhibits the cancer stem cell phenotype and epithelial-to-mesenchymal transition in breast cancer. Cell Cycle 2016; 16:737-745. [PMID: 27753527 DOI: 10.1080/15384101.2016.1241929] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Experimental evidence suggest that breast tumors originate from breast cancer stem cells (BCSCs), and that mitochondrial biogenesis is essential for the anchorage-independent clonal expansion and survival of CSCs, thus rendering mitochondria a significant target for novel treatment approaches. One of the recognized side effects of the FDA-approved drug, doxycycline is the inhibition of mitochondrial biogenesis. Here we investigate the mechanism by which doxycycline exerts its inhibitory effects on the properties of breast cancer cells and BCSCs, such as mammosphere forming efficiency, invasion, migration, apoptosis, the expression of stem cell markers and epithelial-to-mesenchymal transition (EMT) related markers of breast cancer cells. In addition, we explored whether autophagy plays a role in the inhibitory effect of doxycycline on breast cancer cells. We find that doxycyline can inhibit the viability and proliferation of breast cancer cells and BCSCs, decrease mammosphere forming efficiency, migration and invasion, and EMT of breast cancer cells. Expression of stem cell factors Oct4, Sox2, Nanog and CD44 were also significantly downregulated after doxycycline treatment. Moreover, doxycycline could down-regulate the expression of the autophagy marker LC-3BI and LC-3BII, suggesting that inhibiting autophagy may be responsible in part for the observed effects on proliferation, EMT and stem cell markers. The potent inhibition of EMT and cancer stem-like characteristics in breast cancer cells by doxycycline treatment suggests that this drug can be repurposed as an anti-cancer drug in the treatment of breast cancer patients in the clinic.
Collapse
Affiliation(s)
- Le Zhang
- a Department of Oncology , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , P.R. China.,b Department of Radiation Oncology , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Liang Xu
- a Department of Oncology , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , P.R. China.,c Department of Prevention and Cure Center of Breast Disease , Third Hospital of Nanchang , Nanchang , P.R. China
| | - Fengchun Zhang
- a Department of Oncology , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , P.R. China
| | - Erina Vlashi
- b Department of Radiation Oncology , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA.,d Jonsson Comprehensive Cancer Center at UCLA , Los Angeles , CA , USA
| |
Collapse
|
32
|
Aspirin, lysine, mifepristone and doxycycline combined can effectively and safely prevent and treat cancer metastasis: prevent seeds from gemmating on soil. Oncotarget 2016; 6:35157-72. [PMID: 26459390 PMCID: PMC4742096 DOI: 10.18632/oncotarget.6038] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 09/14/2015] [Indexed: 12/11/2022] Open
Abstract
Recent scientific advances have increased our understanding of the cancer metastatic complexities and provided further impetus for new combination therapies to prevent cancer metastasis. Here, we demonstrated that a combination (HAMPT) of aspirin, lysine, mifepristone and doxycycline can effectively and safely prevent cancer metastasis. The pharmaceutically-formulated HAMPT inhibited adhesion of cancer cells to either endothelial cells or extracellular matrix via down-regulating cell adhesion molecules ICAM-1 and α4-integrin. HAMPT inhibited the cloak effect by activated platelets on cancer cells, thereby interfering adhesion and invasion of cancer cells to the underlying stroma. At the effective concentration, HAMPT induced cancer cells into dormancy with minor inhibition on cell viability. Four-day pretreatment followed by 30-day oral administration of HAMPT (33.5-134 mg/kg) to the mice inoculated with cancer cells produced significant inhibition on cancer metastasis dose-dependently without marked side effects. Fifty-day rat toxicity study with HAMPT at doses (335-1340 mg/kg) 20-fold higher than its therapeutic dose produced no significant toxicity. Interestingly, the acute toxic death could not be reached at the maximum administrable dose (5 g/kg). This proof-of-concept study provides the first conceptual evidence that cancer metastasis can be controlled by using affordable old drugs to restrain circulating tumor cells from gemmating on the metastatic soil without the need for cytotoxicity.
Collapse
|
33
|
Qin Y, Zhang Q, Lee S, Zhong WL, Liu YR, Liu HJ, Zhao D, Chen S, Xiao T, Meng J, Jing XS, Wang J, Sun B, Dai TT, Yang C, Sun T, Zhou HG. Doxycycline reverses epithelial-to-mesenchymal transition and suppresses the proliferation and metastasis of lung cancer cells. Oncotarget 2016; 6:40667-79. [PMID: 26512779 PMCID: PMC4747360 DOI: 10.18632/oncotarget.5842] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 09/24/2015] [Indexed: 12/20/2022] Open
Abstract
The gelatinase inhibitor doxycycline is the prototypical antitumor antibiotic. We investigated the effects of doxycycline on the migration, invasion, and metastasis of human lung cancer cell lines and in a mouse model. We also measured the effect of doxycycline on the transcription of epithelial-mesenchymal transition (EMT) markers, and used immunohistochemistry to determine whether EMT reversal was associated with doxycycline inhibition. Doxycycline dose-dependently inhibited proliferation, migration, and invasion of NCI-H446 human small cell lung cancer cells. It also suppressed tumor growth from NCI-H446 and A549 lung cancer cell xenografts without altering body weight, inhibited Lewis lung carcinoma cell migration, and prolonged survival. The activities of the transcription factors Twist1/2, SNAI1/2, AP1, NF-κB, and Stat3 were suppressed by doxycycline, which reversed EMT and inhibited signal transduction, thereby suppressing tumor growth and metastasis. Our data demonstrate functional targeting of transcription factors by doxycycline to reverse EMT and suppress tumor proliferation and metastasis. Thus, doxycycline selectively targets malignant tumors and reduces its metastatic potential with less cytotoxicity in lung cancer patients.
Collapse
Affiliation(s)
- Yuan Qin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Qiang Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Shan Lee
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Wei-Long Zhong
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Yan-Rong Liu
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Hui-Juan Liu
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Dong Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Shuang Chen
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Ting Xiao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Jing Meng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Xue-Shuang Jing
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Jing Wang
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Bo Sun
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Ting-Ting Dai
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Hong-Gang Zhou
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| |
Collapse
|
34
|
Affiliation(s)
- David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | |
Collapse
|
35
|
Nonobe Y, Yokoyama T, Kamikubo Y, Yoshida S, Hisajima N, Shinohara H, Shiraishi Y, Sakurai T, Tabata T. Application of surface plasmon resonance imaging to monitoring G protein-coupled receptor signaling and its modulation in a heterologous expression system. BMC Biotechnol 2016; 16:36. [PMID: 27068216 PMCID: PMC4828886 DOI: 10.1186/s12896-016-0266-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 04/07/2016] [Indexed: 01/30/2023] Open
Abstract
Background G protein-coupled receptors (GPCRs) are ubiquitous surface proteins mediating various biological responses and thus, important targets for therapeutic drugs. GPCRs individually produce their own signaling as well as modulate the signaling of other GPCRs. Real-time observation of GPCR signaling and modulation in living cells is key to molecular study of biological responses and pharmaceutical development. However, fluorescence imaging, the technique widely used for this purpose, requires a fluorescent dye which may inhibit biological responses or a fluorescent-tagged target protein created through time-consuming genetic manipulation. In this study, we applied two-dimensional surface plasmon resonance (SPR) imaging to monitoring the translocation of protein kinase C (PKC), a major GPCR-coupled signaling molecule in the widely used HEK293 cell lines and examined whether the signaling of, and, modulation between heterologously expressed GPCRs can be measured without fluorescent labeling. Results We cultured HEK293 cells on the gold-plated slide glass and evoked SPR at the interface between the cell’s plasma membrane and the gold surface with incident light. The translocation of activated native PKC to the plasma membrane is expected to alter the incident angle-SPR extent relation, and this could be detected as a change in the intensity of light reflection from the specimen illuminated at a fixed incident angle. Direct activation of PKC with 12-O-tetradecanoylphorbol-13-acetate increased the reflection intensity. This increase indeed reported PKC translocation because it was reduced by a pre-treatment with bisindolylmaleimide-1, a PKC inhibitor. We further applied this technique to a stable HEK293 cell line heterologously expressing the GPCRs type-1 metabotropic glutamate receptor (mGluR1) and adenosine A1 receptor (A1R). (RS)-3,5-dihydroxyphenylglycine, a mGluR1 agonist, increased the reflection intensity, and the PKC inhibitor reduced this increase. A pre-treatment with (R)-N6-phenylisopropyladenosine, an A1R-selective agonist suppressed mGluR1-mediated reflection increase. These results suggest that our technique can detect PKC translocation initiated by ligand binding to mGluR1 and its modulation by A1R. Conclusions SPR imaging turned out to be utilizable for monitoring GPCR-mediated PKC translocation and its modulation by a different GPCR in a heterologous expression system. This technique provides a powerful yet easy-to-use tool for molecular study of biological responses and pharmaceutical development.
Collapse
Affiliation(s)
- Yuki Nonobe
- Laboratory for Medical Information Sensing, Graduate School of Science and Engineering, University of Toyama, 3190 Gokufu, Toyama, Toyama, 930-8555, Japan
| | - Tomoki Yokoyama
- Laboratory for Medical Information Sensing, Graduate School of Science and Engineering, University of Toyama, 3190 Gokufu, Toyama, Toyama, 930-8555, Japan
| | - Yuji Kamikubo
- Department of Pharmacology, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Sho Yoshida
- Laboratory for Medical Information Sensing, Graduate School of Science and Engineering, University of Toyama, 3190 Gokufu, Toyama, Toyama, 930-8555, Japan
| | - Nozomi Hisajima
- Laboratory for Medical Information Sensing, Graduate School of Science and Engineering, University of Toyama, 3190 Gokufu, Toyama, Toyama, 930-8555, Japan
| | - Hiroaki Shinohara
- Laboratory for Bioelectronics, Graduate School of Innovative Life Science, University of Toyama, 3190 Gokufu, Toyama, Toyama, 930-8555, Japan
| | - Yuki Shiraishi
- Laboratory for Bioelectronics, Graduate School of Innovative Life Science, University of Toyama, 3190 Gokufu, Toyama, Toyama, 930-8555, Japan
| | - Takashi Sakurai
- Department of Pharmacology, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Toshihide Tabata
- Laboratory for Medical Information Sensing, Graduate School of Science and Engineering, University of Toyama, 3190 Gokufu, Toyama, Toyama, 930-8555, Japan.
| |
Collapse
|
36
|
Ozsvári B, Lamb R, Lisanti MP. Repurposing of FDA-approved drugs against cancer - focus on metastasis. Aging (Albany NY) 2016; 8:567-568. [PMID: 27038638 PMCID: PMC4925811 DOI: 10.18632/aging.100941] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/02/2016] [Indexed: 06/05/2023]
Affiliation(s)
- Béla Ozsvári
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, CRUK Manchester Institute, University of Manchester, Manchester, UK
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, CRUK Manchester Institute, University of Manchester, Manchester, UK
| | - Rebecca Lamb
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, CRUK Manchester Institute, University of Manchester, Manchester, UK
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, CRUK Manchester Institute, University of Manchester, Manchester, UK
| | - Michael P Lisanti
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, CRUK Manchester Institute, University of Manchester, Manchester, UK
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, CRUK Manchester Institute, University of Manchester, Manchester, UK
| |
Collapse
|
37
|
Cirigliano A, Stirpe A, Menta S, Mori M, Dell'Edera D, Pick E, Negri R, Botta B, Rinaldi T. Yeast as a tool to select inhibitors of the cullin deneddylating enzyme Csn5. J Enzyme Inhib Med Chem 2016; 31:1632-7. [PMID: 27028668 DOI: 10.3109/14756366.2016.1160901] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The CSN complex plays a key role in various cellular pathways: through a metalloprotease activity of its Csn5 deneddylating enzyme, it regulates the activity of Cullin-RING ligases (CRLs). Indeed, Csn5 has been found amplified in many tumors, but, due to its pleiotropic effects, it is difficult to dissect its function and the involvement in cancer progression. Moreover, while growing evidences point to the neddylation function as a good target for drug development; specific inhibitors have not yet been developed for the CSN. Here, we propose the yeast Saccharomyces cerevisiae as a model system to screen libraries of small molecules as inhibitors of cullins deneddylation, taking advantage of the unique feature of this organism to survive without a functional CSN5 gene and to accumulate a fully neddylated cullin substrate. By combining molecular modeling and simple genetic tools, we were able to identify two small molecular fragments as selective inhibitors of Csn5 deneddylation function.
Collapse
Affiliation(s)
- Angela Cirigliano
- a Istituto Pasteur Fondazione Cenci Bolognetti, Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy .,b Associazione Gian Franco Lupo "Un sorriso alla vita" Onlus, U.O.D. Laboratorio di Citogenetica e Genetica Molecolare, ASM Matera , Italy
| | - Alessandro Stirpe
- a Istituto Pasteur Fondazione Cenci Bolognetti, Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy
| | - Sergio Menta
- c Dipartimento di Chimica e Tecnologie del Farmaco , Sapienza University of Rome , Rome , Italy
| | - Mattia Mori
- d Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia , Rome , Italy , and
| | - Domenico Dell'Edera
- b Associazione Gian Franco Lupo "Un sorriso alla vita" Onlus, U.O.D. Laboratorio di Citogenetica e Genetica Molecolare, ASM Matera , Italy
| | - Elah Pick
- e Department of Biology and Environment , Faculty of Natural Sciences, University of Haifa , Oranim , Kiryat Tivon , Israel
| | - Rodolfo Negri
- a Istituto Pasteur Fondazione Cenci Bolognetti, Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy
| | - Bruno Botta
- c Dipartimento di Chimica e Tecnologie del Farmaco , Sapienza University of Rome , Rome , Italy
| | - Teresa Rinaldi
- a Istituto Pasteur Fondazione Cenci Bolognetti, Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy
| |
Collapse
|
38
|
New Insights Into the Mechanism of COP9 Signalosome-Cullin-RING Ubiquitin-Ligase Pathway Deregulation in Urological Cancers. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 323:181-229. [PMID: 26944622 DOI: 10.1016/bs.ircmb.2015.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Urological cancers are a very common type of cancer worldwide and have alarming high incidence and mortality rates, especially in kidney cancers, illustrate the urgent need for new therapeutic targets. Recent publications point to a deregulated COP9 signalosome (CSN)-cullin-RING ubiquitin-ligase (CRL) pathway which is here considered and investigated as potential target in urological cancers with strong focus on renal cell carcinomas (RCC). The CSN forms supercomplexes with CRLs in order to preserve protein homeostasis and was found deregulated in several cancer types. Examination of selected CSN-CRL pathway components in RCC patient samples and four RCC cell lines revealed an interesting deregulated p27(Kip1)-Skp2-CAND1 axis and two p27(Kip1) point mutations in 786-O cells; p27(Kip1)V109G and p27(Kip1)I119T. The p27(Kip1) mutants were detected in patients with RCC and appear to be responsible for an accelerated growth rate in 786-O cells. The occurrence of p27(Kip1)V109G and p27(Kip1)I119T in RCC makes the CSN-CRL pathway an attractive therapeutic target.
Collapse
|
39
|
Peiris-Pagès M, Sotgia F, Lisanti MP. Doxycycline and therapeutic targeting of the DNA damage response in cancer cells: old drug, new purpose. Oncoscience 2015; 2:696-9. [PMID: 26425660 PMCID: PMC4580062 DOI: 10.18632/oncoscience.215] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 08/17/2015] [Indexed: 12/19/2022] Open
Abstract
There is a small proportion of cells within a tumour with self-renewing properties, which is resistant to conventional therapy, and is responsible for tumour initiation, maintenance and metastasis. These cells are known as cancer stem cells (CSCs) or tumour-initiating cells (TICs) [1]. Recent publications identify several antibiotics, such as salinomycin or doxycycline, as selective CSCs inhibitors [2-4]. However, the mechanisms of action of these antibiotics on CSCs are not fully understood.
Collapse
Affiliation(s)
- Maria Peiris-Pagès
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester, UK ; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, Manchester, UK
| | - Federica Sotgia
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester, UK ; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, Manchester, UK
| | - Michael P Lisanti
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester, UK ; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|