1
|
Wang Y, Hsu P, Hu H, Lin F, Wei X. Role of arachidonic acid metabolism in osteosarcoma prognosis by integrating WGCNA and bioinformatics analysis. BMC Cancer 2025; 25:445. [PMID: 40075313 PMCID: PMC11905593 DOI: 10.1186/s12885-024-13278-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/02/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Osteosarcoma is a rare tumor with poor clinical outcomes. New therapeutic targets are urgently needed. Previous research indicates that genes abnormally expressed in osteosarcoma are significantly involved in the arachidonic acid (AA) metabolic pathway. However, the role of arachidonic acid metabolism-related genes (AAMRGs) in osteosarcoma prognosis remains unknown. METHODS Osteosarcoma samples from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were classified into high-score and low-score groups based on AAMRGs scores obtained through ssGSEA analysis. The intersecting genes were identified from weighted gene co-expression network analysis (WGCNA), DEGs (osteosarcoma vs. normal) and DE-AAMRGs (high- vs. low-score). An AA metabolism predictive model of the five AAMRGs were established by Cox regression and the LASSO algorithm. Model performance was evaluated using Kaplan-Meier survival and receiver operating characteristic (ROC) curve analysis. In vitro experiments of the AA related biomarkers was validated. RESULTS Our study constructed an AAMRGs prognostic signature (CD36, CLDN11, STOM, EPYC, PANX3). K-M analysis indicated that patients in the low-risk group showed superior overall survival to high-risk group (p<0.05). ROC curves showed that all AUC values in the prognostic model exceeded 0.76. By ESTIMATE algorithms, we discovered that patients in high-risk groups had lower immune score, stromal score, and estimate score. Correlation analysis showed the strongest positive correlation between STOM and natural killer cells, and the highest negative association between PANX3 and central memory CD8 T cells. An AAMRGs prognostic signature was constructed for osteosarcoma prognosis. CONCLUSION The study suggested that a high level of AAMRGs might serve as a biomarker for poor prognosis in osteosarcoma and offers a potential explanation for the role of cyclooxygenase inhibitors in cancer. The five biomarkers (CD36, CLDN11, EPYC, PANX3, and STOM) were screened to construct an AAMRGs risk model with prognostic value, providing a new reference for the prognosis and treatment of osteosarcoma.
Collapse
Affiliation(s)
- Yaling Wang
- Department of Oncology, Shanghai Eighth People's Hospital, Shanghai, China
| | - Peichun Hsu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Hu
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Lin
- Department of Oncology, Shanghai Eighth People's Hospital, Shanghai, China.
| | - Xiaokang Wei
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Ducatez F, Berger MG, Pilon C, Plichet T, Lesueur C, Berger J, Belmatoug N, Marret S, Bekri S, Tebani A. Deciphering metabolic shifts in Gaucher disease type 1: a multi-omics study. J Mol Med (Berl) 2025; 103:187-203. [PMID: 39738845 DOI: 10.1007/s00109-024-02512-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/13/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
Gaucher disease (GD), an autosomal recessive lysosomal disorder, primarily affects the lysosomal enzyme β-glucocerebrosidase (GCase), leading to glucosylceramide accumulation in lysosomes. GD presents a wide spectrum of clinical manifestations. This study deploys immune-based proteomics and mass spectrometry-based metabolomics technologies to comprehensively investigate the biochemical landscape in 43 deeply phenotyped type 1 GD patients compared to 59 controls. Conventional and systems biology approaches have been used to analyze the data. The results show promising biological imprints. Elevated phosphatidylcholines in GD patients suggest altered lipid metabolism, potentially due to their increased synthesis. This points to endoplasmic reticulum stress and impaired lipid trafficking, commonly seen in lysosomal diseases. GD patients exhibit an inflammatory profile with elevated cytokines and autoimmune-like inflammation, even in treated patients, highlighting the complexity of GD-related immune imbalances. Mitochondrial dysfunction clues are found through increased oxidative stress markers and altered acylcarnitine profiles in GD patients, suggesting mitochondrial membrane dysfunction affecting carnitine-carrying capacity. Furthermore, platelet count, splenectomy, treatment, and clinical traits were associated with specific omics features, providing insights into GD's clinical heterogeneity and potential diagnostic markers. Autophagy inhibition appears pivotal in GD, driving lipid synthesis, impaired mitochondrial function, and inflammation through chronic activation of mTORC1. Despite limitations like focusing on type 1 GD and using targeted omics approaches, this study provides valuable insights into GD metabolic and immune dysregulation. It lays the basis for future comprehensive investigations into GD manifestations with broader scope and molecular coverage. KEY MESSAGES: The study sheds light on metabolic and immune dysregulation in Gaucher disease. Gaucher disease patients showed elevated phosphatidylcholines, disrupted lipid metabolism, and inflammation profiles. Signs of mitochondrial dysfunction are evident in Gaucher disease patients, with autophagy inhibition significantly affecting lipid synthesis, mitochondrial function, and inflammation via chronic activation of mTORC1.
Collapse
Affiliation(s)
- Franklin Ducatez
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Marc G Berger
- CHU Clermont-Ferrand, Hopital Estaing, CRB-Auvergne, 63003, Clermont-Ferrand, France
- Université Clermont Auvergne, EA, 7453 CHELTER, 63000, Clermont-Ferrand, France
| | - Carine Pilon
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Thomas Plichet
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Céline Lesueur
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Juliette Berger
- CHU Clermont-Ferrand, Hopital Estaing, CRB-Auvergne, 63003, Clermont-Ferrand, France
- Université Clermont Auvergne, EA, 7453 CHELTER, 63000, Clermont-Ferrand, France
| | - Nadia Belmatoug
- Referral Center for Lysosomal Diseases, Filière G2M, Paris Cité University, APHP-Nord, Paris, France
| | - Stéphane Marret
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Abdellah Tebani
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France.
| |
Collapse
|
3
|
Ashcroft FJ, Bourboula A, Mahammad N, Barbayianni E, Feuerherm AJ, Nguyen TT, Hayashi D, Kokotou MG, Alevizopoulos K, Dennis EA, Kokotos G, Johansen B. Next generation thiazolyl ketone inhibitors of cytosolic phospholipase A 2 α for targeted cancer therapy. Nat Commun 2025; 16:164. [PMID: 39747052 PMCID: PMC11696576 DOI: 10.1038/s41467-024-55536-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Eicosanoids are key players in inflammatory diseases and cancer. Targeting their production by inhibiting Group IVA cytosolic phospholipase A2 (cPLA2α) offers a promising approach for cancer therapy. In this study, we synthesize a second generation of thiazolyl ketone inhibitors of cPLA2α starting with compound GK470 (AVX235) and test their in vitro and cellular activities. We identify a more potent and selective lead molecule, GK420 (AVX420), which we test in parallel with AVX235 and a structurally unrelated compound, AVX002 for inhibition of cell viability across a panel of cancer cell lines. From this, we show that activity of polycomb group repressive complex 2 is a key molecular determinant of sensitivity to cPLA2α inhibition, while resistance depends on antioxidant response pathways. Consistent with these results, we show that elevated intracellular reactive oxygen species and activating transcription factor 4 target gene expression precede cell death in AVX420-sensitive T-cell acute lymphoblastic leukemia cells. Our findings imply cPLA2α may support cancer by mitigating oxidative stress and inhibiting tumor suppressor expression and suggest that AVX420 has potential for treating acute leukemias and other cancers that are susceptible to oxidative cell death.
Collapse
Affiliation(s)
- Felicity J Ashcroft
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Asimina Bourboula
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Nur Mahammad
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Efrosini Barbayianni
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Astrid J Feuerherm
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Thanh Thuy Nguyen
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Daiki Hayashi
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Maroula G Kokotou
- Laboratory of Chemistry, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| | | | - Edward A Dennis
- Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, CA, USA
- Department of Pharmacology, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece.
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece.
| | - Berit Johansen
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
4
|
Pearce SM, Cross NA, Smith DP, Clench MR, Flint LE, Hamm G, Goodwin R, Langridge JI, Claude E, Cole LM. Multimodal Mass Spectrometry Imaging of an Osteosarcoma Multicellular Tumour Spheroid Model to Investigate Drug-Induced Response. Metabolites 2024; 14:315. [PMID: 38921450 PMCID: PMC11205347 DOI: 10.3390/metabo14060315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
A multimodal mass spectrometry imaging (MSI) approach was used to investigate the chemotherapy drug-induced response of a Multicellular Tumour Spheroid (MCTS) 3D cell culture model of osteosarcoma (OS). The work addresses the critical demand for enhanced translatable early drug discovery approaches by demonstrating a robust spatially resolved molecular distribution analysis in tumour models following chemotherapeutic intervention. Advanced high-resolution techniques were employed, including desorption electrospray ionisation (DESI) mass spectrometry imaging (MSI), to assess the interplay between metabolic and cellular pathways in response to chemotherapeutic intervention. Endogenous metabolite distributions of the human OS tumour models were complemented with subcellularly resolved protein localisation by the detection of metal-tagged antibodies using Imaging Mass Cytometry (IMC). The first application of matrix-assisted laser desorption ionization-immunohistochemistry (MALDI-IHC) of 3D cell culture models is reported here. Protein localisation and expression following an acute dosage of the chemotherapy drug doxorubicin demonstrated novel indications for mechanisms of region-specific tumour survival and cell-cycle-specific drug-induced responses. Previously unknown doxorubicin-induced metabolite upregulation was revealed by DESI-MSI of MCTSs, which may be used to inform mechanisms of chemotherapeutic resistance. The demonstration of specific tumour survival mechanisms that are characteristic of those reported for in vivo tumours has underscored the increasing value of this approach as a tool to investigate drug resistance.
Collapse
Affiliation(s)
- Sophie M. Pearce
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Neil A. Cross
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - David P. Smith
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Malcolm R. Clench
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Lucy E. Flint
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - Gregory Hamm
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - Richard Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - James I. Langridge
- Waters Corporation, Stamford Avenue, Altrincham Road, Wilmslow, Cheshire SK9 4AX, UK; (J.I.L.); (E.C.)
| | - Emmanuelle Claude
- Waters Corporation, Stamford Avenue, Altrincham Road, Wilmslow, Cheshire SK9 4AX, UK; (J.I.L.); (E.C.)
| | - Laura M. Cole
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| |
Collapse
|
5
|
Benzo Y, Prada JG, Dattilo MA, Bigi MM, Castillo AF, Mori Sequeiros Garcia MM, Poderoso C, Maloberti PM. Acyl-CoA synthetase 4 modulates mitochondrial function in breast cancer cells. Heliyon 2024; 10:e30639. [PMID: 38756582 PMCID: PMC11096749 DOI: 10.1016/j.heliyon.2024.e30639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Mitochondria are dynamic organelles that respond to cellular stress through changes in global mass, interconnection, and subcellular location. As mitochondria play an important role in tumor development and progression, alterations in energy metabolism allow tumor cells to survive and spread even in challenging conditions. Alterations in mitochondrial bioenergetics have been recently proposed as a hallmark of cancer, and positive regulation of lipid metabolism constitutes one of the most common metabolic changes observed in tumor cells. Acyl-CoA synthetase 4 (ACSL4) is an enzyme catalyzing the activation of long chain polyunsaturated fatty acids with a strong substrate preference for arachidonic acid (AA). High ACSL4 expression has been related to aggressive cancer phenotypes, including breast cancer, and its overexpression has been shown to positively regulate the mammalian Target of Rapamycin (mTOR) pathway, involved in the regulation of mitochondrial metabolism genes. However, little is known about the role of ACSL4 in the regulation of mitochondrial function and metabolism in cancer cells. In this context, our objective was to study whether mitochondrial function and metabolism, processes usually altered in tumors, are modulated by ACSL4 in breast cancer cells. Using ACSL4 overexpression in MCF-7 cells, we demonstrate that this enzyme can increase the mRNA and protein levels of essential mitochondrial regulatory proteins such as nuclear respiratory factor 1 (NRF-1), voltage-dependent anion channel 1 (VDAC1) and respiratory chain Complex III. Furthermore, respiratory parameters analysis revealed an increase in oxygen consumption rate (OCR) and in spare respiratory capacity (SRC), among others. ACSL4 knockdown in MDA-MB-231 cells led to the decrease in OCR and in SCR, supporting the role of ACSL4 in the regulation of mitochondrial bioenergetics. Moreover, ACSL4 overexpression induced an increase in glycolytic function, in keeping with an increase in mitochondrial respiratory activity. Finally, there was a decrease in mitochondrial mass detected in cells that overexpressed ACSL4, while the knockdown of ACSL4 expression in MDA-MB-231 cells showed the opposite effect. Altogether, these results unveil the role of ACSL4 in mitochondrial function and metabolism and expand the knowledge of ACSL4 participation in pathological processes such as breast cancer.
Collapse
Affiliation(s)
- Yanina Benzo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Jesica G. Prada
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Melina A. Dattilo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - María Mercedes Bigi
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Ana F. Castillo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - María Mercedes Mori Sequeiros Garcia
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Cecilia Poderoso
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Paula M. Maloberti
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| |
Collapse
|
6
|
Asiamah EA, Feng B, Guo R, Yaxing X, Du X, Liu X, Zhang J, Cui H, Ma J. The Contributions of the Endolysosomal Compartment and Autophagy to APOEɛ4 Allele-Mediated Increase in Alzheimer's Disease Risk. J Alzheimers Dis 2024; 97:1007-1031. [PMID: 38306054 DOI: 10.3233/jad-230658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Apolipoprotein E4 (APOE4), although yet-to-be fully understood, increases the risk and lowers the age of onset of Alzheimer's disease (AD), which is the major cause of dementia among elderly individuals. The endosome-lysosome and autophagy pathways, which are necessary for homeostasis in both neurons and glia, are dysregulated even in early AD. Nonetheless, the contributory roles of these pathways to developing AD-related pathologies in APOE4 individuals and models are unclear. Therefore, this review summarizes the dysregulations in the endosome-lysosome and autophagy pathways in APOE4 individuals and non-human models, and how these anomalies contribute to developing AD-relevant pathologies. The available literature suggests that APOE4 causes endosomal enlargement, increases endosomal acidification, impairs endosomal recycling, and downregulates exosome production. APOE4 impairs autophagy initiation and inhibits basal autophagy and autophagy flux. APOE4 promotes lysosome formation and trafficking and causes ApoE to accumulate in lysosomes. APOE4-mediated changes in the endosome, autophagosome and lysosome could promote AD-related features including Aβ accumulation, tau hyperphosphorylation, glial dysfunction, lipid dyshomeostasis, and synaptic defects. ApoE4 protein could mediate APOE4-mediated endosome-lysosome-autophagy changes. ApoE4 impairs vesicle recycling and endosome trafficking, impairs the synthesis of autophagy genes, resists being dissociated from its receptors and degradation, and forms a stable folding intermediate that could disrupt lysosome structure. Drugs such as molecular correctors that target ApoE4 molecular structure and enhance autophagy may ameliorate the endosome-lysosome-autophagy-mediated increase in AD risk in APOE4 individuals.
Collapse
Affiliation(s)
- Ernest Amponsah Asiamah
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, PMB UCC, Cape Coast, Ghana
| | - Baofeng Feng
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei, China
| | - Ruiyun Guo
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Xu Yaxing
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Xiaofeng Du
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Xin Liu
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Jinyu Zhang
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Huixian Cui
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei, China
| | - Jun Ma
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei, China
| |
Collapse
|
7
|
Santos-Pereira M, Pereira SC, Rebelo I, Spadella MA, Oliveira PF, Alves MG. Decoding the Influence of Obesity on Prostate Cancer and Its Transgenerational Impact. Nutrients 2023; 15:4858. [PMID: 38068717 PMCID: PMC10707940 DOI: 10.3390/nu15234858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/12/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
In recent decades, the escalating prevalence of metabolic disorders, notably obesity and being overweight, has emerged as a pressing concern in public health. Projections for the future indicate a continual upward trajectory in obesity rates, primarily attributable to unhealthy dietary patterns and sedentary lifestyles. The ramifications of obesity extend beyond its visible manifestations, intricately weaving a web of hormonal dysregulation, chronic inflammation, and oxidative stress. This nexus of factors holds particular significance in the context of carcinogenesis, notably in the case of prostate cancer (PCa), which is a pervasive malignancy and a leading cause of mortality among men. A compelling hypothesis arises from the perspective of transgenerational inheritance, wherein genetic and epigenetic imprints associated with obesity may wield influence over the development of PCa. This review proposes a comprehensive exploration of the nuanced mechanisms through which obesity disrupts prostate homeostasis and serves as a catalyst for PCa initiation. Additionally, it delves into the intriguing interplay between the transgenerational transmission of both obesity-related traits and the predisposition to PCa. Drawing insights from a spectrum of sources, ranging from in vitro and animal model research to human studies, this review endeavors to discuss the intricate connections between obesity and PCa. However, the landscape remains partially obscured as the current state of knowledge unveils only fragments of the complex mechanisms linking these phenomena. As research advances, unraveling the associated factors and underlying mechanisms promises to unveil novel avenues for understanding and potentially mitigating the nexus between obesity and the development of PCa.
Collapse
Affiliation(s)
- Mariana Santos-Pereira
- iBiMED-Institute of Biomedicine and Department of Medical Science, University of Aveiro, 3810-193 Aveiro, Portugal;
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal;
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal
| | - Sara C. Pereira
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal;
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal
- LAQV-REQUIMTE and Department of Chemistry, Campus Universitario de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Irene Rebelo
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biologic Sciences, Pharmaceutical Faculty, University of Porto, 4050-313 Porto, Portugal;
| | - Maria A. Spadella
- Human Embryology Laboratory, Marília Medical School, Marília 17519-030, SP, Brazil;
| | - Pedro F. Oliveira
- LAQV-REQUIMTE and Department of Chemistry, Campus Universitario de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Marco G. Alves
- iBiMED-Institute of Biomedicine and Department of Medical Science, University of Aveiro, 3810-193 Aveiro, Portugal;
| |
Collapse
|
8
|
Asleh K, Dery V, Taylor C, Davey M, Djeungoue-Petga MA, Ouellette RJ. Extracellular vesicle-based liquid biopsy biomarkers and their application in precision immuno-oncology. Biomark Res 2023; 11:99. [PMID: 37978566 PMCID: PMC10655470 DOI: 10.1186/s40364-023-00540-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
While the field of precision oncology is rapidly expanding and more targeted options are revolutionizing cancer treatment paradigms, therapeutic resistance particularly to immunotherapy remains a pressing challenge. This can be largely attributed to the dynamic tumor-stroma interactions that continuously alter the microenvironment. While to date most advancements have been made through examining the clinical utility of tissue-based biomarkers, their invasive nature and lack of a holistic representation of the evolving disease in a real-time manner could result in suboptimal treatment decisions. Thus, using minimally-invasive approaches to identify biomarkers that predict and monitor treatment response as well as alert to the emergence of recurrences is of a critical need. Currently, research efforts are shifting towards developing liquid biopsy-based biomarkers obtained from patients over the course of disease. Liquid biopsy represents a unique opportunity to monitor intercellular communication within the tumor microenvironment which could occur through the exchange of extracellular vesicles (EVs). EVs are lipid bilayer membrane nanoscale vesicles which transfer a plethora of biomolecules that mediate intercellular crosstalk, shape the tumor microenvironment, and modify drug response. The capture of EVs using innovative approaches, such as microfluidics, magnetic beads, and aptamers, allow their analysis via high throughput multi-omics techniques and facilitate their use for biomarker discovery. Artificial intelligence, using machine and deep learning algorithms, is advancing multi-omics analyses to uncover candidate biomarkers and predictive signatures that are key for translation into clinical trials. With the increasing recognition of the role of EVs in mediating immune evasion and as a valuable biomarker source, these real-time snapshots of cellular communication are promising to become an important tool in the field of precision oncology and spur the recognition of strategies to block resistance to immunotherapy. In this review, we discuss the emerging role of EVs in biomarker research describing current advances in their isolation and analysis techniques as well as their function as mediators in the tumor microenvironment. We also highlight recent lung cancer and melanoma studies that point towards their application as predictive biomarkers for immunotherapy and their potential clinical use in precision immuno-oncology.
Collapse
Affiliation(s)
- Karama Asleh
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada.
| | - Valerie Dery
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
| | - Catherine Taylor
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Michelle Davey
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | | | - Rodney J Ouellette
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- Dr Georges L. Dumont University Hospital, Vitalite Health Network, Moncton, New Brunswick, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| |
Collapse
|
9
|
Hautanen V, Toimela T, Paparella M, Heinonen T. A Human Cell-based Assay to Assess the Induction of Vasculature Formation for Non-genotoxic Carcinogenicity Testing Purposes: A Pilot Study. Altern Lab Anim 2023:2611929231171165. [PMID: 37125451 DOI: 10.1177/02611929231171165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The induction of vasculature formation is proposed to be a significant mechanism behind the non-genotoxic carcinogenicity of a chemical. The vasculature formation model used in this study is based on the coculture of human primary HUVECs and hASCs. This model was used to develop an assay to assess the induction of vasculature formation. Three assay protocols, based on different conditions, were developed and compared in order to identify the optimal conditions required. Some serum supplements and growth factors were observed to be essential for initiating vasculature formation. Of the studied putative positive reference chemicals, aspartame, sodium nitrite, bisphenol A and nicotine treatment led to a clear induction of vasculature formation, but arsenic and cadmium treatment only led to a slight increase. This human cell-based assay has the potential to be used as one test within a next generation testing battery, to assess the non-genotoxic carcinogenicity of a chemical through the mechanism of vasculature formation induction.
Collapse
Affiliation(s)
- Veera Hautanen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tarja Toimela
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Martin Paparella
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Tuula Heinonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
10
|
Khan SA, Ilies MA. The Phospholipase A2 Superfamily: Structure, Isozymes, Catalysis, Physiologic and Pathologic Roles. Int J Mol Sci 2023; 24:ijms24021353. [PMID: 36674864 PMCID: PMC9862071 DOI: 10.3390/ijms24021353] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/23/2022] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
The phospholipase A2 (PLA2) superfamily of phospholipase enzymes hydrolyzes the ester bond at the sn-2 position of the phospholipids, generating a free fatty acid and a lysophospholipid. The PLA2s are amphiphilic in nature and work only at the water/lipid interface, acting on phospholipid assemblies rather than on isolated single phospholipids. The superfamily of PLA2 comprises at least six big families of isoenzymes, based on their structure, location, substrate specificity and physiologic roles. We are reviewing the secreted PLA2 (sPLA2), cytosolic PLA2 (cPLA2), Ca2+-independent PLA2 (iPLA2), lipoprotein-associated PLA2 (LpPLA2), lysosomal PLA2 (LPLA2) and adipose-tissue-specific PLA2 (AdPLA2), focusing on the differences in their structure, mechanism of action, substrate specificity, interfacial kinetics and tissue distribution. The PLA2s play important roles both physiologically and pathologically, with their expression increasing significantly in diseases such as sepsis, inflammation, different cancers, glaucoma, obesity and Alzheimer's disease, which are also detailed in this review.
Collapse
|
11
|
Helfenberger KE, Argentino GF, Benzo Y, Herrera LM, Finocchietto P, Poderoso C. Angiotensin II Regulates Mitochondrial mTOR Pathway Activity Dependent on Acyl-CoA Synthetase 4 in Adrenocortical Cells. Endocrinology 2022; 163:6763139. [PMID: 36256598 DOI: 10.1210/endocr/bqac170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Indexed: 11/19/2022]
Abstract
Two well-known protein complexes in mammalian cells, mTOR type 1 and type 2 (mTORC1/2) are involved in several cellular processes such as protein synthesis, cell proliferation, and commonly dysregulated in cancer. An acyl-CoA synthetase type 4 (ACSL4) is one of the most recently mTORC1/2 regulators described, in breast cancer cells. The expression of ACSL4 is hormone-regulated in adrenocortical cells and required for steroid biosynthesis. mTORC1/2 have been reported to be crucial in the proliferation of human adrenocortical tumor cells H295R and interestingly reported at several subcellular locations, which has brought cell biology to the vanguard of the mTOR signaling field. In the present work, we study the regulation of mTORC1/2 activation by angiotensin II (Ang II)-the trophic hormone for adrenocortical cells-the subcellular localization of mTORC1/2 signaling proteins and the role of ACSL4 in the regulation of this pathway, in H295R cells. Ang II promotes activation by phosphorylation of mTORC1/2 pathway proteins in a time-dependent manner. Mitochondrial pools of ribosomal protein S6, protein kinase B (Akt) in threonine 308, and serine 473 and Rictor are phosphorylated and activated. Glycogen synthase kinase type 3 (GSK3) is phosphorylated and inactivated in mitochondria, favoring mTORC1 activation. Epidermal growth factor, a classic mTORC1/2 activator, promoted unique activation kinetics of mTORC1/2 pathway, except for Akt phosphorylation. Here, we demonstrate that ACSL4 is necessary for mTORC1/2 effectors phosphorylation and H295R proliferation, triggered by Ang II. Ang II promotes activation of mitochondrial mTORC1/2 signaling proteins, through ACSL4, with a direct effect on adrenocortical cellular proliferation.
Collapse
Affiliation(s)
- Katia E Helfenberger
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Giuliana F Argentino
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Yanina Benzo
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Lucía M Herrera
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Paola Finocchietto
- Laboratorio del Metabolismo del Oxígeno. Hospital de Clínicas "José de San Martín," Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Cecilia Poderoso
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| |
Collapse
|
12
|
Abstract
The mechanistic target of the rapamycin (mTOR) signaling pathway is the central regulator of cell growth and proliferation by integrating growth factor and nutrient availability. Under healthy physiological conditions, this process is tightly coordinated and essential to maintain whole-body homeostasis. Not surprisingly, dysregulated mTOR signaling underpins several diseases with increasing incidence worldwide, including obesity, diabetes, and cancer. Consequently, there is significant clinical interest in developing therapeutic strategies that effectively target this pathway. The transition of mTOR inhibitors from the bench to bedside, however, has largely been marked with challenges and shortcomings, such as the development of therapy resistance and adverse side effects in patients. In this review, we discuss the current status of first-, second-, and third-generation mTOR inhibitors as a cancer therapy in both preclinical and clinical settings, with a particular emphasis on the mechanisms of drug resistance. We focus especially on the emerging role of diet as an important environmental determinant of therapy response, and posit a conceptual framework that links nutrient availability and whole-body metabolic states such as obesity with many of the previously defined processes that drive resistance to mTOR-targeted therapies. Given the role of mTOR as a central integrator of cell metabolism and function, we propose that modulating nutrient inputs through dietary interventions may influence the signaling dynamics of this pathway and compensatory nodes. In doing so, new opportunities for exploiting diet/drug synergies are highlighted that may unlock the therapeutic potential of mTOR inhibitors as a cancer treatment.
Collapse
Affiliation(s)
- Nikos Koundouros
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021,USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA
- Correspondence: Nikos Koundouros, Meyer Cancer Center, Weill Cornell Medicine, 413 East 69th Street, New York, NY, 10021 USA.
| | - John Blenis
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021,USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
- Correspondence: John Blenis, Meyer Cancer Center, Weill Cornell Medicine, 413 East 69th Street, New York, NY, 10021 USA.
| |
Collapse
|
13
|
Pournajaf S, Dargahi L, Javan M, Pourgholami MH. Molecular Pharmacology and Novel Potential Therapeutic Applications of Fingolimod. Front Pharmacol 2022; 13:807639. [PMID: 35250559 PMCID: PMC8889014 DOI: 10.3389/fphar.2022.807639] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Fingolimod is a well-tolerated, highly effective disease-modifying therapy successfully utilized in the management of multiple sclerosis. The active metabolite, fingolimod-phosphate, acts on sphingosine-1-phosphate receptors (S1PRs) to bring about an array of pharmacological effects. While being initially recognized as a novel agent that can profoundly reduce T-cell numbers in circulation and the CNS, thereby suppressing inflammation and MS, there is now rapidly increasing knowledge on its previously unrecognized molecular and potential therapeutic effects in diverse pathological conditions. In addition to exerting inhibitory effects on sphingolipid pathway enzymes, fingolimod also inhibits histone deacetylases, transient receptor potential cation channel subfamily M member 7 (TRMP7), cytosolic phospholipase A2α (cPLA2α), reduces lysophosphatidic acid (LPA) plasma levels, and activates protein phosphatase 2A (PP2A). Furthermore, fingolimod induces apoptosis, autophagy, cell cycle arrest, epigenetic regulations, macrophages M1/M2 shift and enhances BDNF expression. According to recent evidence, fingolimod modulates a range of other molecular pathways deeply rooted in disease initiation or progression. Experimental reports have firmly associated the drug with potentially beneficial therapeutic effects in immunomodulatory diseases, CNS injuries, and diseases including Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, and even cancer. Attractive pharmacological effects, relative safety, favorable pharmacokinetics, and positive experimental data have collectively led to its testing in clinical trials. Based on the recent reports, fingolimod may soon find its way as an adjunct therapy in various disparate pathological conditions. This review summarizes the up-to-date knowledge about molecular pharmacology and potential therapeutic uses of fingolimod.
Collapse
Affiliation(s)
- Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
14
|
Dey S, Singh AK, Singh AK, Rawat K, Banerjee J, Agnihotri V, Upadhaya D. Critical pathways of oral squamous cell carcinoma: molecular biomarker and therapeutic intervention. Med Oncol 2022; 39:30. [DOI: 10.1007/s12032-021-01633-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
|
15
|
Harmati M, Bukva M, Böröczky T, Buzás K, Gyukity-Sebestyén E. The role of the metabolite cargo of extracellular vesicles in tumor progression. Cancer Metastasis Rev 2021; 40:1203-1221. [PMID: 34957539 PMCID: PMC8825386 DOI: 10.1007/s10555-021-10014-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022]
Abstract
Metabolomic reprogramming in tumor and stroma cells is a hallmark of cancer but understanding its effects on the metabolite composition and function of tumor-derived extracellular vesicles (EVs) is still in its infancy. EVs are membrane-bound sacs with a complex molecular composition secreted by all living cells. They are key mediators of intercellular communication both in normal and pathological conditions and play a crucial role in tumor development. Although lipids are major components of EVs, most of the EV cargo studies have targeted proteins and nucleic acids. The potential of the EV metabolome as a source for biomarker discovery has gained recognition recently, but knowledge on the biological activity of tumor EV metabolites still remains limited. Therefore, we aimed (i) to compile the list of metabolites identified in tumor EVs isolated from either clinical specimens or in vitro samples and (ii) describe their role in tumor progression through literature search and pathway analysis.
Collapse
Affiliation(s)
- Mária Harmati
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre - Eötvös Loránd Research Network, 6726, Szeged, Hungary
| | - Mátyás Bukva
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre - Eötvös Loránd Research Network, 6726, Szeged, Hungary.,Department of Immunology, University of Szeged, 6720, Szeged, Hungary.,Doctoral School of Interdisciplinary Medicine, University of Szeged, 6720, Szeged, Hungary
| | - Tímea Böröczky
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre - Eötvös Loránd Research Network, 6726, Szeged, Hungary.,Department of Immunology, University of Szeged, 6720, Szeged, Hungary.,Doctoral School of Interdisciplinary Medicine, University of Szeged, 6720, Szeged, Hungary
| | - Krisztina Buzás
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre - Eötvös Loránd Research Network, 6726, Szeged, Hungary.,Department of Immunology, University of Szeged, 6720, Szeged, Hungary
| | - Edina Gyukity-Sebestyén
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre - Eötvös Loránd Research Network, 6726, Szeged, Hungary.
| |
Collapse
|
16
|
Sun Q, Zhao H, Liu Z, Wang F, He Q, Xiu C, Guo L, Tian Q, Fan L, Sun J, Sun D. Identifying potential metabolic tissue biomarkers for papillary thyroid cancer in different iodine nutrient regions. Endocrine 2021; 74:582-591. [PMID: 34075541 DOI: 10.1007/s12020-021-02773-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/19/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE To investigate the applicability of metabolomics to select thyroid cancer-associated biomarkers and discover the effects of iodine on metabolic changes in thyroid cancer. METHODS In this study, a total of 33 papillary thyroid cancer (PTC) patients from areas with iodine excess and 32 PTC patients from areas with adequate iodine were recruited, and their cancerous tissue and paracancerous tissue were collected. These specimens were analyzed by ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC/QTOF/MS) in conjunction with multivariate statistical analysis. RESULTS Good separations were obtained for PTC tissue vs. paracancerous tissue, and 15 metabolites, including L-octanoylcarnitine, N-arachidonoylglycine, and others were found to be disturbed in PTC tissue. Moreover, the metabolic profile presented considerable separation between PTC tissue from different iodine areas, and 15 metabolomic biomarkers were found to be differentially expressed. Among them, 10 metabolites, including arachidonoylcarnitine and LysoPCs, were related to thyroid cancer and excess iodine. These biomarkers play a role in arachidonic acid metabolism pathways and others. In addition, biomarkers such as 3,5-tetradecadiencarnitine and oxidized glutathione were significantly correlated with thyroid function, and biomarkers such as L-octanoylcarnitine and arachidonic acid were significantly correlated with the clinical characteristics of PTC. CONCLUSIONS Distinct differences in metabolic profiles were found to exist between PTCs from areas with different levels of iodine nutrition. The identified biomarkers have significant potential for diagnosing PTC and investigating its underlying mechanisms.
Collapse
Affiliation(s)
- Qihao Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongjian Zhao
- General Surgery Department, People's Hospital of Chengwu County, Heze, Shandong, China
| | - Zhiyong Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China
| | - Fengqian Wang
- Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Qian He
- Shandong First Medical University, Tai'an, Shandong, China
| | - Cheng Xiu
- Department of Head and Neck Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lunhua Guo
- Department of Head and Neck Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qiushi Tian
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China
| | - Lijun Fan
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China.
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.
| | - Ji Sun
- Department of Head and Neck Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
17
|
Alblihy A, Shoqafi A, Toss MS, Algethami M, Harris AE, Jeyapalan JN, Abdel-Fatah T, Servante J, Chan SYT, Green A, Mongan NP, Rakha EA, Madhusudan S. Untangling the clinicopathological significance of MRE11-RAD50-NBS1 complex in sporadic breast cancers. NPJ Breast Cancer 2021; 7:143. [PMID: 34782604 PMCID: PMC8593132 DOI: 10.1038/s41523-021-00350-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 10/22/2021] [Indexed: 12/27/2022] Open
Abstract
The MRE11-RAD50-NBS1 (MRN) complex is critical for genomic stability. Although germline mutations in MRN may increase breast cancer susceptibility, such mutations are extremely rare. Here, we have conducted a comprehensive clinicopathological study of MRN in sporadic breast cancers. We have protein expression profiled for MRN and a panel of DNA repair factors involved in double-strand break repair (BRCA1, BRCA2, ATM, CHK2, ATR, Chk1, pChk1, RAD51, γH2AX, RPA1, RPA2, DNA-PKcs), RECQ DNA helicases (BLM, WRN, RECQ1, RECQL4, RECQ5), nucleotide excision repair (ERCC1) and base excision repair (SMUG1, APE1, FEN1, PARP1, XRCC1, Pol β) in 1650 clinical breast cancers. The prognostic significance of MRE11, RAD50 and NBS1 transcripts and their microRNA regulators (hsa-miR-494 and hsa-miR-99b) were evaluated in large clinical datasets. Expression of MRN components was analysed in The Cancer Genome Atlas breast cancer cohort. We show that low nuclear MRN is linked to aggressive histopathological phenotypes such as high tumour grade, high mitotic index, oestrogen receptor- and high-risk Nottingham Prognostic Index. In univariate analysis, low nuclear MRE11 and low nuclear RAD50 were associated with poor survival. In multivariate analysis, low nuclear RAD50 remained independently linked with adverse clinical outcomes. Low RAD50 transcripts were also linked with reduced survival. In contrast, overexpression of hsa-miR-494 and hsa-miR-99b microRNAs was associated with poor survival. We observed large-scale genome-wide alterations in MRN-deficient tumours contributing to aggressive behaviour. We conclude that MRN status may be a useful tool to stratify tumours for precision medicine strategies.
Collapse
Affiliation(s)
- Adel Alblihy
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK
- Medical Center, King Fahad Security College (KFSC), Riyadh, 11461, Saudi Arabia
| | - Ahmed Shoqafi
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK
| | - Michael S Toss
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK
- Department of Pathology, Nottingham University Hospitals, City Hospital Campus, Nottingham, NG5 1PB, UK
| | - Mashael Algethami
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK
| | - Anna E Harris
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK
| | - Jennie N Jeyapalan
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK
| | - Tarek Abdel-Fatah
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham, NG5 1PB, UK
| | | | - Stephen Y T Chan
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham, NG5 1PB, UK
| | - Andrew Green
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK
| | - Nigel P Mongan
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK
| | - Emad A Rakha
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK
- Department of Pathology, Nottingham University Hospitals, City Hospital Campus, Nottingham, NG5 1PB, UK
| | - Srinivasan Madhusudan
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 3RD, UK.
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham, NG5 1PB, UK.
| |
Collapse
|
18
|
Emerging role of ferroptosis in breast cancer: New dawn for overcoming tumor progression. Pharmacol Ther 2021; 232:107992. [PMID: 34606782 DOI: 10.1016/j.pharmthera.2021.107992] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer has become a serious threat to women's health. Cancer progression is mainly derived from resistance to apoptosis induced by procedures or therapies. Therefore, new drugs or models that can overcome apoptosis resistance should be identified. Ferroptosis is a recently identified mode of cell death characterized by excess reactive oxygen species-induced lipid peroxidation. Since ferroptosis is distinct from apoptosis, necrosis and autophagy, its induction successfully eliminates cancer cells that are resistant to other modes of cell death. Therefore, ferroptosis may become a new direction around which to design breast cancer treatment. Unfortunately, the complete appearance of ferroptosis in breast cancer has not yet been fully elucidated. Furthermore, whether ferroptosis inducers can be used in combination with traditional anti- breast cancer drugs is still unknown. Moreover, a summary of ferroptosis in breast cancer progression and therapy is currently not available. In this review, we discuss the roles of ferroptosis-associated modulators glutathione, glutathione peroxidase 4, iron, nuclear factor erythroid-2 related factor-2, superoxide dismutases, lipoxygenase and coenzyme Q in breast cancer. Furthermore, we provide evidence that traditional drugs against breast cancer induce ferroptosis, and that ferroptosis inducers eliminate breast cancer cells. Finally, we put forward prospect of using ferroptosis inducers in breast cancer therapy, and predict possible obstacles and corresponding solutions. This review will deepen our understanding of the relationship between ferroptosis and breast cancer, and provide new insights into breast cancer-related therapeutic strategies.
Collapse
|
19
|
Mukerjee S, Saeedan AS, Ansari MN, Singh M. Polyunsaturated Fatty Acids Mediated Regulation of Membrane Biochemistry and Tumor Cell Membrane Integrity. MEMBRANES 2021; 11:479. [PMID: 34203433 PMCID: PMC8304949 DOI: 10.3390/membranes11070479] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/05/2021] [Accepted: 06/24/2021] [Indexed: 12/25/2022]
Abstract
Particular dramatic macromolecule proteins are responsible for various cellular events in our body system. Lipids have recently recognized a lot more attention of scientists for understanding the relationship between lipid and cellular function and human health However, a biological membrane is formed with a lipid bilayer, which is called a P-L-P design. Our body system is balanced through various communicative signaling pathways derived from biological membrane proteins and lipids. In the case of any fatal disease such as cancer, the biological membrane compositions are altered. To repair the biological membrane composition and prevent cancer, dietary fatty acids, such as omega-3 polyunsaturated fatty acids, are essential in human health but are not directly synthesized in our body system. In this review, we will discuss the alteration of the biological membrane composition in breast cancer. We will highlight the role of dietary fatty acids in altering cellular composition in the P-L-P bilayer. We will also address the importance of omega-3 polyunsaturated fatty acids to regulate the membrane fluidity of cancer cells.
Collapse
Affiliation(s)
- Souvik Mukerjee
- Department of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur 495009, Chhattisgarh, India;
| | - Abdulaziz S. Saeedan
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Mohd. Nazam Ansari
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Manjari Singh
- Department of Pharmaceutical Sciences, Assam University, Silchar 788011, Assam, India
| |
Collapse
|
20
|
Chen X, Chen X, Sun X, Wang C, Wen Z, Cheng Y. RAD001 targeted HUVECs reverses 12-lipoxygenase-induced angiogenesis in oesophageal squamous cell carcinoma. J Cell Mol Med 2021; 25:6936-6947. [PMID: 34120414 PMCID: PMC8278093 DOI: 10.1111/jcmm.16705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/11/2021] [Accepted: 05/18/2021] [Indexed: 12/28/2022] Open
Abstract
12‐LOX plays an important role in the progression of various malignancies. However, the underlying mechanisms of the action of 12‐LOX and tumour treatment strategies remain not fully defined. In this study, we investigated the possible roles of 12‐LOX in ESCC and explored the new therapeutic target. Approximately 73% of ESCC tissues showed marked up‐regulation of 12‐LOX, which was associated with poor prognosis. 12‐LOX overexpression was positively correlated with the malignant progression of ESCC as demonstrated both in vitro and in vivo. Up‐regulation of 12‐LOX significantly increased the proliferation of ESCC cells and the xenograft volume. Moreover, 12‐LOX up‐regulation promoted tube formation of HUVECs and tumour angiogenesis in xenografts. Mechanism investigation indicated that 12‐LOX overexpression led to activation of the PI3K/AKT/mTOR pathway and the up‐regulation of VEGF in ESCC cells. Subsequent analysis indicated that the RAD001 could reverse the 12‐LOX‐induced promoting effect on ESCC. Specifically, the application of RAD001 inhibited the proliferation of ESCC cells and the tube‐forming ability of HUVECs. In the drug group, the xenografts exhibited significant volume reduction and angiogenesis inhibition. We demonstrated that RAD001 could inhibit HUVEC migration. These findings presented the evidence that RAD001 had distinct roles on HUVECs and could exert anti‐tumour effects by targeting not only the PI3K/AKT/mTOR pathway but the angiogenesis in ESCC.
Collapse
Affiliation(s)
- Xue Chen
- Departments of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Xuan Chen
- Departments of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaozheng Sun
- Departments of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Cong Wang
- Departments of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhihua Wen
- Departments of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Yufeng Cheng
- Departments of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
21
|
Lu J, Guan H, Wu D, Hu Z, Zhang H, Jiang H, Yu J, Zeng K, Li H, Zhang H, Pan C, Cai D, Yu X. Pseudolaric acid B ameliorates synovial inflammation and vessel formation by stabilizing PPARγ to inhibit NF-κB signalling pathway. J Cell Mol Med 2021; 25:6664-6678. [PMID: 34117708 PMCID: PMC8278075 DOI: 10.1111/jcmm.16670] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/10/2021] [Accepted: 05/08/2021] [Indexed: 12/16/2022] Open
Abstract
Synovial macrophage polarization and inflammation are essential for osteoarthritis (OA) development, yet the molecular mechanisms and regulation responsible for the pathogenesis are still poorly understood. Here, we report that pseudolaric acid B (PAB) attenuated articular cartilage degeneration and synovitis during OA. PAB, a diterpene acid, specifically inhibited NF-κB signalling and reduced the production of pro-inflammatory cytokines, which further decreased M1 polarization and vessel formation. We further provide in vivo and in vitro evidences that PAB suppressed NF-κB signalling by stabilizing PPARγ. Using PPARγ antagonist could abolish anti-inflammatory effect of PAB and rescue the activation of NF-κB signalling during OA. Our findings identify a previously unrecognized role of PAB in the regulation of OA and provide mechanisms by which PAB regulates NF-κB signalling through PPARγ, which further suggest targeting synovial inflammation or inhibiting vessel formation at early stage could be an effective preventive strategy for OA.
Collapse
Affiliation(s)
- Jiansen Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Joint Surgery, the Fifth Affiliated Hospital of Southern Medical University Guangdong Province, Guangzhou, China
| | - Hong Guan
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.,Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhiqiang Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongbo Zhang
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.,Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Huaji Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingyao Yu
- School of Basic Medical Sciences, Gannan Medical University, Ganzhou, China
| | - Ke Zeng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongyu Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Haiyan Zhang
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.,Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Chenglong Pan
- Department of Joint Surgery, the Fifth Affiliated Hospital of Southern Medical University Guangdong Province, Guangzhou, China
| | - Daozhang Cai
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.,Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Joint Surgery, the Fifth Affiliated Hospital of Southern Medical University Guangdong Province, Guangzhou, China.,Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Bustamante-Marin XM, Merlino JL, Devericks E, Carson MS, Hursting SD, Stewart DA. Mechanistic Targets and Nutritionally Relevant Intervention Strategies to Break Obesity-Breast Cancer Links. Front Endocrinol (Lausanne) 2021; 12:632284. [PMID: 33815289 PMCID: PMC8011316 DOI: 10.3389/fendo.2021.632284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/17/2021] [Indexed: 12/29/2022] Open
Abstract
The worldwide prevalence of overweight and obesity has tripled since 1975. In the United States, the percentage of adults who are obese exceeds 42.5%. Individuals with obesity often display multiple metabolic perturbations, such as insulin resistance and persistent inflammation, which can suppress the immune system. These alterations in homeostatic mechanisms underlie the clinical parameters of metabolic syndrome, an established risk factor for many cancers, including breast cancer. Within the growth-promoting, proinflammatory milieu of the obese state, crosstalk between adipocytes, immune cells and breast epithelial cells occurs via obesity-associated hormones, angiogenic factors, cytokines, and other mediators that can enhance breast cancer risk and/or progression. This review synthesizes evidence on the biological mechanisms underlying obesity-breast cancer links, with emphasis on emerging mechanism-based interventions in the context of nutrition, using modifiable elements of diet alone or paired with physical activity, to reduce the burden of obesity on breast cancer.
Collapse
Affiliation(s)
| | - Jenna L. Merlino
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - Emily Devericks
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - Meredith S. Carson
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC, United States
| | - Delisha A. Stewart
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC, United States
| |
Collapse
|
23
|
Bandyopadhayaya S, Akimov MG, Verma R, Sharma A, Sharma D, Kundu GC, Gretskaya NM, Bezuglov VV, Mandal CC. N-arachidonoyl dopamine inhibits epithelial-mesenchymal transition of breast cancer cells through ERK signaling and decreasing the cellular cholesterol. J Biochem Mol Toxicol 2021; 35:e22693. [PMID: 33393692 DOI: 10.1002/jbt.22693] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/04/2020] [Accepted: 12/11/2020] [Indexed: 01/02/2023]
Abstract
N-acyl dopamines (NADAs) are bioactive lipids of the endovanilloid family with known cytotoxicity for the cancer cells; however, the available data on the participation of the endovanilloids in epithelial-mesenchymal transition (EMT) and cancer stemness are controversial. This study unveils the inhibitory role of N-arachidonoyl dopamine (AA-DA), a typical representative of the NADA family, in breast cancer cell migration, EMT, and stemness. AA-DA treatment also led to a decrease in cholesterol biosynthesis gene expressions, and addition of exogenous cholesterol reverted these AA-DA-mediated inhibitory effects. Notably, AA-DA treatment inhibited the key regulatory gene of the cholesterol biosynthesis pathway, sterol regulatory element-binding protein 1 (SREBP1), with concurrent repression of the endoplasmic reticulum kinase 1/2 (ERK1/2) pathway. Furthermore, U0126, an ERK inhibitor, inhibited SREBP1 and decreased cellular cholesterol level, unwinding the molecular mechanism behind AA-DA-mediated anticancer activity. Thus, we, for the first time, revealed that AA-DA counteracts breast cancer EMT via inhibition of ERK signaling and cholesterol content.
Collapse
Affiliation(s)
- Shreetama Bandyopadhayaya
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Mikhail G Akimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS, Moscow, Russia
| | - Ranjeet Verma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Ankit Sharma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Divya Sharma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Gopal C Kundu
- School of Biotechnology, Institute of Eminence, KIIT Deemed to be University, Bhubaneswar, India
| | - Natalia M Gretskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS, Moscow, Russia
| | - Vladimir V Bezuglov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS, Moscow, Russia
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
24
|
FADS1 promotes the progression of laryngeal squamous cell carcinoma through activating AKT/mTOR signaling. Cell Death Dis 2020; 11:272. [PMID: 32332698 PMCID: PMC7181692 DOI: 10.1038/s41419-020-2457-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Metabolic abnormality is the major feature of laryngeal squamous cell carcinoma (LSCC), however, the underlying mechanism remain largely elusive. Fatty acid desaturase 1 (FADS1), as the key rate-limiting enzyme of polyunsaturated fatty acids (PUFAs), catalyzes dihomo-gamma-linolenic acid (DGLA) to arachidonic acid (AA). In this study, we reported that the expression of FADS1 was upregulated in LSCC, high FADS1 expression was closely associated with the advanced clinical features and poor prognosis of the recurrent LSCC patients after chemotherapy. Liquid chromatograph-mass spectrometry (LC-MS) analysis revealed that FADS1 overexpression induced greater conversion of DGLA to AA, suggesting an increased activity of FADS1. Similarly, the level of prostaglandin E2 (PGE2), a downstream metabolite of AA, was also elevated in cancerous laryngeal tissues. Functional assays showed that FADS1 knockdown suppressed the proliferation, migration and invasion of LSCC cells, while FADS1 overexpression had the opposite effects. Bioinformatic analysis based on microarray data found that FADS1 could activate AKT/mTOR signaling. This hypothesis was further validated by both in vivo and in vitro assays. Hence, our data has supported the viewpoint that FADS1 is a potential promoter in LSCC progression, and has laid the foundation for further functional research on the PUFA dietary supplementation interventions targeting FADS1/AKT/mTOR pathway for LSCC prevention and treatment.
Collapse
|
25
|
Tunset HM, Feuerherm AJ, Selvik LKM, Johansen B, Moestue SA. Cytosolic Phospholipase A2 Alpha Regulates TLR Signaling and Migration in Metastatic 4T1 Cells. Int J Mol Sci 2019; 20:ijms20194800. [PMID: 31569627 PMCID: PMC6801560 DOI: 10.3390/ijms20194800] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/14/2019] [Indexed: 12/02/2022] Open
Abstract
Metastatic disease is the leading cause of death in breast cancer patients. Disrupting the cancer cell’s ability to migrate may be a strategy for hindering metastasis. Cytosolic phospholipase A2 α (cPLA2α), along with downstream proinflammatory and promigratory metabolites, has been implicated in several aspects of tumorigenesis, as well as metastasis, in various types of cancer. In this study, we aim to characterize the response to reduced cPLA2α activity in metastatic versus non-metastatic cells. We employ an isogenic murine cell line pair displaying metastatic (4T1) and non-metastatic (67NR) phenotype to investigate the role of cPLA2α on migration. Furthermore, we elucidate the effect of reduced cPLA2α activity on global gene expression in the metastatic cell line. Enzyme inhibition is achieved by using a competitive pharmacological inhibitor, cPLA2α inhibitor X (CIX). Our data show that 4T1 expresses significantly higher cPLA2α levels as compared to 67NR, and the two cell lines show different sensitivity to the CIX treatment with regards to metabolism and proliferation. Inhibition of cPLA2α at nontoxic concentrations attenuates migration of highly metastatic 4T1 cells, but not non-metastatic 67NR cells. Gene expression analysis indicates that processes such as interferon type I (IFN-I) signaling and cell cycle regulation are key processes regulated by cPLA2a in metastatic 4T1 cells, supporting the findings from the biological assays. This study demonstrates that two isogenic cancer cell lines with different metastatic potential respond differently to reduced cPLA2α activity. In conclusion, we argue that cPLA2α is a potential therapeutic target in cancer and that enzyme inhibition may inhibit metastasis through an anti-migratory mechanism, possibly involving Toll-like receptor signaling and type I interferons.
Collapse
Affiliation(s)
- Hanna Maja Tunset
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491 Trondheim, Norway.
| | - Astrid Jullumstrø Feuerherm
- Center for Oral Health Services and Research (TkMidt), 7030 Trondheim, Norway.
- Department of Biology, Norwegian University of Science and Technology, Realfagbygget, 7491 Trondheim, Norway.
| | - Linn-Karina Myrland Selvik
- Department of Biology, Norwegian University of Science and Technology, Realfagbygget, 7491 Trondheim, Norway.
| | - Berit Johansen
- Department of Biology, Norwegian University of Science and Technology, Realfagbygget, 7491 Trondheim, Norway.
| | - Siver Andreas Moestue
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491 Trondheim, Norway.
- Department of Health Sciences, Nord University, P.O. Box 1490, 8049 Bodø, Norway.
| |
Collapse
|
26
|
Metabolomics profiling of haloperidol and validation of thromboxane-related signaling in the early development of zebrafish. Biochem Biophys Res Commun 2019; 513:608-615. [PMID: 30981506 DOI: 10.1016/j.bbrc.2019.04.003] [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] [Received: 03/22/2019] [Accepted: 04/01/2019] [Indexed: 01/30/2023]
Abstract
Haloperidol is a common butyrophenone-derivative antipsychotic drug that is used clinically to treat schizophrenia and to control Tourette disorder. Haloperidol has been shown to be an embryonic toxicant and to cause a variety of adverse effects that affect human embryonic development. However, the pathway impaired by haloperidol during the developmental stages remains unclear. To elucidate the innate toxicological pathway of haloperidol, we investigated the lethality of haloperidol during the embryonic development of zebrafish. We observed that haloperidol caused serious morphological changes, with an LD50 of 9.7 x 10-6 ± 2.4 x 10-6 μg/L. Next, we established a systematic approach to perform metabolite profiling in embryonic zebrafish with various concentrations of haloperidol and analyzed the metabolites using ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC/Q-TOF MS). A total of 304 metabolites were identified and 86 metabolites were chosen to predict potential pathways. Among the metabolites, we found through prediction that numerous metabolomics-biological pathways are associated with haloperidol, including peroxisome-proliferator-activated receptor (ppar), thromboxane, and mTOR signaling. Quantitative real time-qPCR was then used to validate the gene expression potentially associated with the thromboxane, which is a metabolic product of arachidonic acid and considered to be important for cell proliferation and the inflammatory response. To sum up, analysis of metabolites in the zebrafish model provides a system for mining biomarkers that reflect biological significance and highlight the therapeutic potency in humans. In addition, it may show potential for application to other pharmaceuticals to identify their various activities and clarify functional mechanisms in the future.
Collapse
|
27
|
Pang X, Yin P, Han J, Wang Z, Zheng F, Chen X. cPLA 2a correlates with metastasis and poor prognosis of osteosarcoma by facilitating epithelial-mesenchymal transition. Pathol Res Pract 2019; 215:152398. [PMID: 31003849 DOI: 10.1016/j.prp.2019.03.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/11/2019] [Accepted: 03/26/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Osteosarcoma (OS) patients with metastasis have very dismal prognoses, and lack effective target therapies. Overexpression of cytosolic phospholipase A2 (cPLA2) has been shown to promote progression in several types of cancers, but its functions in OS have not been investigated. MATERIALS AND METHODS In our study, the expression of cPLA2a was detected with immunohistochemistry in 102 cases of OS. The clinical significance of cPLA2a was evaluated by analyzing its correlation with clinicopathological factors. The prognostic significance of cPLA2a was estimated by univariate and multivariate analysis. The oncogenic functions of cPLA2a on cell proliferation and invasion were investigated by MTT assay and tranwell assay respectively. Western blotting was applied to detect the markers of epithelial-mesenchymal transition (EMT) after silencing cPLA2a expression or inhibiting its activity by a specific antagonist. RESULTS In our study, high expression of cPLA2a was significantly associated with metastasis and advanced Enneking stage. High cPLA2a expression was significantly associated with poor prognosis and it was an independent prognostic biomarker of OS. By silencing cPLA2a or inhibiting its activity by a specific antagonist, we demonstrated that cPLA2a promoted cell invasion of OS cells via inducing the EMT process. CONCLUSIONS High cPLA2a expression was an independent prognostic biomarker of OS, and cPLA2a could promote OS cell invasion via inducing the EMT process, indicating that cPLA2a was an independent prognostic biomarker and may be an effective drug target for OS.
Collapse
Affiliation(s)
- Xumei Pang
- Department of Oncology, Yidu Central Hospital of Weifang City, Weifang, China
| | - Peng Yin
- Department of Orthopedic Surgery, Yidu Central Hospital of Weifang City, Weifang, China
| | - Jiliang Han
- Department of Radiotherapy, Yidu Central Hospital of Weifang City, Weifang, China
| | - Zhiqian Wang
- Department of Oncology, Yidu Central Hospital of Weifang City, Weifang, China
| | - Feng Zheng
- Department of Orthopedics, The Affiliated Hospital of Putian University, Putian, Fujian, China
| | - Xuanhuang Chen
- Department of Orthopedics, The Affiliated Hospital of Putian University, Putian, Fujian, China.
| |
Collapse
|
28
|
Canoy JL, Bitacura JG. Cytotoxicity and Antiangiogenic Activity of Turbinaria ornata Agardh and Padina australis Hauck Ethanolic Extracts. Anal Cell Pathol (Amst) 2018; 2018:3709491. [PMID: 30159217 PMCID: PMC6109477 DOI: 10.1155/2018/3709491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/29/2018] [Accepted: 07/03/2018] [Indexed: 01/24/2023] Open
Abstract
Brown macroalgae species are constantly reported as potential sources of bioactive compounds useful in inhibiting cell proliferation and vascular formation. Thus, this study was conducted to determine and compare the in vitro cytotoxic activities of Turbinaria ornata Agardh and Padina australis Hauck ethanolic extracts against baker's yeast (Saccharomyces cerevisiae) using the resazurin reduction test (RRT) and investigate their in vivo antiangiogenic activity through duck (Anas platyrhynchos) chorioallantoic membrane (CAM) assay. Both T. ornata and P. australis ethanolic extracts exhibited cytotoxic activities at IC50 of 530.53 ppm and 528.78 ppm, respectively, and significant cytotoxicity was determined in 750 ppm and 1000 ppm concentrations of T. ornata and 1000 ppm concentration of P. australis. Also, both T. ornata and P. australis ethanolic extracts exhibited antiangiogenic activity (100% vascular inhibition) as all the concentrations of both species caused severe vascular damage in all the duck CAM samples treated. These results show the potential future application of these species for cytotoxic activities and vascular inhibition. The conduct of further tests using other model systems is recommended.
Collapse
Affiliation(s)
- Jenefa L. Canoy
- Department of Biological Sciences, Visayas State University, Visca, 6521-A Baybay City, Leyte, Philippines
| | - Jayzon G. Bitacura
- Department of Biological Sciences, Visayas State University, Visca, 6521-A Baybay City, Leyte, Philippines
| |
Collapse
|
29
|
Expression of Cytosolic Phospholipase A2 Alpha in Glioblastoma Is Associated With Resistance to Chemotherapy. Am J Med Sci 2018; 356:391-398. [PMID: 30360807 DOI: 10.1016/j.amjms.2018.06.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 06/17/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND The clinical management of glioblastoma is still challenging despite aggressive surgery and radio-chemotherapy approaches. Better understanding of the molecules involved in glioblastoma chemoresistance is necessary to improve the treatment and predict prognosis. MATERIALS AND METHODS We analyzed the expression and possible roles of cytosolic phospholipase A2 alpha (cPLA2α) in human glioblastoma cell lines and patient samples using immunohistochemistry and cellular assays. We analyzed the signaling pathways that cPLA2α regulates in glioblastoma cells using western blot analysis. RESULTS Our work demonstrated that cPLA2α is upregulated in glioblastoma compared with normal neuron cells. The expression of cPLA2α varies in multiple glioblastoma cell lines and is associated with chemoresistance rather than tumor development. cPLA2α depletion moderately inhibits glioblastoma growth and survival but remarkably sensitizes chemo-resistant glioblastoma cells to several chemotherapeutic agents. Mechanistically, cPLA2α knockdown significantly suppresses the PI3K/Akt/mTOR pathway in glioblastoma cells. CONCLUSIONS We are the first to identify the important role of cPLA2α in glioblastoma in response to chemotherapy. Our data also suggest that cPLA2α may serve as a biomarker to indicate prognosis of glioblastoma patients with high level of cPLA2α to chemotherapy.
Collapse
|
30
|
Nosaka T, Baba T, Tanabe Y, Sasaki S, Nishimura T, Imamura Y, Yurino H, Hashimoto S, Arita M, Nakamoto Y, Mukaida N. Alveolar Macrophages Drive Hepatocellular Carcinoma Lung Metastasis by Generating Leukotriene B 4. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:1839-1852. [PMID: 29378914 DOI: 10.4049/jimmunol.1700544] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 12/29/2017] [Indexed: 11/19/2022]
Abstract
Macrophages in lungs can be classified into two subpopulations, alveolar macrophages (AMs) and interstitial macrophages (IMs), which reside in the alveolar and interstitial spaces, respectively. Accumulating evidence indicates the involvement of IMs in lung metastasis, but the roles of AMs in lung metastasis still remain elusive. An i.v. injection of a mouse hepatocellular carcinoma (HCC) cell line, BNL, caused lung metastasis foci with infiltration of AMs and IMs. Comprehensive determination of arachidonic acid metabolite levels revealed increases in leukotrienes and PGs in lungs in this metastasis model. A 5-lipoxygenase (LOX) inhibitor but not a cyclooxygenase inhibitor reduced the numbers of metastatic foci, particularly those of a larger size. A major 5-LOX metabolite, LTB4, augmented in vitro cell proliferation of human HCC cell lines as well as BNL cells. Moreover, in this lung metastasis course, AMs exhibited higher expression levels of the 5-LOX and LTB4 than IMs. Consistently, 5-LOX-expressing AMs increased in the lungs of human HCC patients with lung metastasis, compared with those without lung metastasis. Furthermore, intratracheal clodronate liposome injection selectively depleted AMs but not IMs, together with reduced LTB4 content and metastatic foci numbers in this lung metastasis process. Finally, IMs in mouse metastatic foci produced CCL2, thereby recruiting blood-borne, CCR2-expressing AMs into lungs. Thus, AMs can be recruited under the guidance of IM-derived CCL2 into metastatic lungs and can eventually contribute to the progression of lung metastasis by providing a potent arachidonic acid-derived tumor growth promoting mediator, LTB4.
Collapse
Affiliation(s)
- Takuto Nosaka
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui 910-1193, Japan
| | - Tomohisa Baba
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan;
| | - Yamato Tanabe
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
| | - Soichiro Sasaki
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Tatsunori Nishimura
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yoshiaki Imamura
- Division of Surgical Pathology, University of Fukui, Yoshida-gun, Fukui 910-1193, Japan
| | - Hideaki Yurino
- Division of Nephrology, Department of Laboratory Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Shinichi Hashimoto
- Division of Nephrology, Department of Laboratory Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
- Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Chiyoda-ku, Tokyo 102-8666, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa 236-0027, Japan; and
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo 108-8345, Japan
| | - Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui 910-1193, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| |
Collapse
|
31
|
Liu M, Zhou L, Zhang B, He M, Dong X, Lin X, Jia C, Bai X, Dai Y, Su Y, Zou Z, Zheng H. Elevation of n-3/n-6 PUFAs ratio suppresses mTORC1 and prevents colorectal carcinogenesis associated with APC mutation. Oncotarget 2018; 7:76944-76954. [PMID: 27769066 PMCID: PMC5363561 DOI: 10.18632/oncotarget.12759] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 10/10/2016] [Indexed: 11/25/2022] Open
Abstract
Although epidemiological and preclinical studies have shown the preventative effect of n-3 polyunsaturated fatty acids (PUFAs) on colorectal cancer (CRC), the underlying molecular mechanisms are not clear. In this study, we revealed that elevation of n-3/n-6 PUFAs ratio suppress the mechanistic target of rapamycin complex 1 (mTORC1) and prevent colorectal tumorigenesis. The transgenic expression of fat-1, a desaturase that catalyzes the conversion of n-6 to n-3 PUFAs and produces n-3 PUFAs endogenously, repressed colorectal tumor cell growth and remarkably reduced tumor burden, and alleviated anemia as well as hyperlipidemia in APCMin/+ (adenomatous polyposis coli) mice, a classic CRC model that best simulates most clinical cases. In contrast to arachidonic acid (AA, C20:4 n-6), either Docosahexaenoic acid (DHA, C22:6 n-3), eicosapentaenoic acid (EPA, C20:5 n-3), or a combination of DHA and AA, efficiently inhibited the proliferation of CRC cell lines and promoted apoptosis in these cells. The ectopic expression of fat-1 had similar effects in colon epithelial cells with APC depletion. Mechanistically, elevation of n-3/n-6 ratio suppressed mTORC1 activity in tumors of APCMin/+ mice, CRC cell lines with APC mutation, and in normal colon epithelial cells with APC depletion. In addition, elevation of n-3/n-6 ratio repressed mTORC1 activity and inhibited adipogenic differentiation in preadipocytes with APC knockdown, as well as alleviated hyperlipidemia in APCMin/+ mice. Taken together, our findings have provided novel insights into the potential mechanism by which increase in n-3/n-6 PUFAs ratio represses CRC development, and also a new rationale for utilizing n-3 PUFAs in CRC prevention and treatment.
Collapse
Affiliation(s)
- Miao Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ling Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Baiyu Zhang
- Department of Rheumatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, 510655, China
| | - Minhong He
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoying Dong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaojun Lin
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Chunhong Jia
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Yifan Dai
- State Key Laboratory of Reproductive Medicine and Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 201129, China
| | - Yongchun Su
- Department of Bioinformatics, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Zhipeng Zou
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Hang Zheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
32
|
Nie J, Chen J, Yang J, Pei Q, Li J, Liu J, Xu L, Li N, Chen Y, Chen X, Luo H, Sun T. Inhibition of mammalian target of rapamycin complex 1 signaling by n-3 polyunsaturated fatty acids promotes locomotor recovery after spinal cord injury. Mol Med Rep 2018; 17:5894-5902. [PMID: 29436695 PMCID: PMC5866035 DOI: 10.3892/mmr.2018.8583] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 01/09/2018] [Indexed: 01/06/2023] Open
Abstract
The present study aimed to explore the effects of n‑3 polyunsaturated fatty acids (PUFAs) on autophagy and their potential for promoting locomotor recovery after spinal cord injury (SCI). Primary neurons were isolated and cultured. Sprague‑Dawley rats were randomly divided into three groups and fed diets with different amounts of n‑3 PUFAs. A model of spinal cord contusion was created at the T10 spinal segment and the composition of PUFAs was analyzed using gas chromatography. Spinal repair and motor function were evaluated postoperatively. Assessment of the effects of n‑3 PUFAs on autophagy and mammalian target of rapamycin complex 1 (mTORC1) was performed using immunofluorescence staining and western blotting. In vitro, n‑3 PUFAs inhibited mTORC1 and enhanced autophagy. The n‑3 PUFA levels and the ratio of n‑3 PUFA to n‑6 PUFA in the spinal cord and serum of rats fed a high‑n‑3 PUFA diet were higher before and after operation (P<0.05). Additionally, rats in the high‑n‑3 PUFA group showed improved motor function recovery, spinal cord repair‑related protein expression level (MBP, Galc and GFAP). Expression levels if these protiens in the high‑n‑3 PUFA diet group expressed the highest levels, followed by the low‑n‑3 PUFA diet group and finally the control group (P<0.05). high‑n‑3 PUFA diet promoted autophagy ability and inhibited activity of the mTORC1 signaling pathway compared with the low‑n‑3 PUFA diet group or the control group (P<0.05). These results suggest that exogenous dietary n‑3 PUFAs can inhibit mTORC1 signaling and enhance autophagy, promoting functional recovery of rats with SCI.
Collapse
Affiliation(s)
- Jiping Nie
- Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jian Chen
- Department of Orthopedics, Three Gorges Central Hospital of Chongqing, Chongqing 404000, P.R. China
| | - Jianguo Yang
- Department of Orthopedics, Huhhot First Hospital, Huhhot, Inner Mongolia Autonomous Region 010020, P.R. China
| | - Qinqin Pei
- Department of Orthopedics, Three Gorges Central Hospital of Chongqing, Chongqing 404000, P.R. China
| | - Jing Li
- Department of Orthopedics, Three Gorges Central Hospital of Chongqing, Chongqing 404000, P.R. China
| | - Jia Liu
- Department of Orthopedics, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Lixin Xu
- Department of Orthopedics, Three Gorges Central Hospital of Chongqing, Chongqing 404000, P.R. China
| | - Nan Li
- Department of Orthopedics, Three Gorges Central Hospital of Chongqing, Chongqing 404000, P.R. China
| | - Youhao Chen
- Department of Orthopedics, Three Gorges Central Hospital of Chongqing, Chongqing 404000, P.R. China
| | - Xiaohua Chen
- Department of Orthopedics, Three Gorges Central Hospital of Chongqing, Chongqing 404000, P.R. China
| | - Hao Luo
- Department of Orthopedics, Three Gorges Central Hospital of Chongqing, Chongqing 404000, P.R. China
| | - Tiansheng Sun
- Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
33
|
Endothelial Ca 2+ Signaling and the Resistance to Anticancer Treatments: Partners in Crime. Int J Mol Sci 2018; 19:ijms19010217. [PMID: 29324706 PMCID: PMC5796166 DOI: 10.3390/ijms19010217] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 02/06/2023] Open
Abstract
Intracellular Ca2+ signaling drives angiogenesis and vasculogenesis by stimulating proliferation, migration, and tube formation in both vascular endothelial cells and endothelial colony forming cells (ECFCs), which represent the only endothelial precursor truly belonging to the endothelial phenotype. In addition, local Ca2+ signals at the endoplasmic reticulum (ER)-mitochondria interface regulate endothelial cell fate by stimulating survival or apoptosis depending on the extent of the mitochondrial Ca2+ increase. The present article aims at describing how remodeling of the endothelial Ca2+ toolkit contributes to establish intrinsic or acquired resistance to standard anti-cancer therapies. The endothelial Ca2+ toolkit undergoes a major alteration in tumor endothelial cells and tumor-associated ECFCs. These include changes in TRPV4 expression and increase in the expression of P2X7 receptors, Piezo2, Stim1, Orai1, TRPC1, TRPC5, Connexin 40 and dysregulation of the ER Ca2+ handling machinery. Additionally, remodeling of the endothelial Ca2+ toolkit could involve nicotinic acetylcholine receptors, gasotransmitters-gated channels, two-pore channels and Na⁺/H⁺ exchanger. Targeting the endothelial Ca2+ toolkit could represent an alternative adjuvant therapy to circumvent patients' resistance to current anti-cancer treatments.
Collapse
|
34
|
Liu F, Pan Z, Zhang J, Ni J, Wang C, Wang Z, Gu F, Dong W, Zhou W, Liu H. Overexpression of RHEB is associated with metastasis and poor prognosis in hepatocellular carcinoma. Oncol Lett 2018; 15:3838-3845. [PMID: 29467900 DOI: 10.3892/ol.2018.7759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/25/2016] [Indexed: 12/11/2022] Open
Abstract
Aberrant expression of Ras homolog enriched in brain (RHEB) has been observed in a variety of cancer tissues and is closely associated with clinicopathological features. However, the expression profile of RHEB in patients with hepatocellular carcinoma (HCC) and its clinical signature with underlying mechanisms have not been explored thus far. To analyze the association between RHEB expression and clinicopathological features, the RHEB expression levels were determined in the present study using gene microarrays, immunohistochemistry and western blotting in 60 liver cancer tissues and 35 normal liver tissues. Downregulation of RHEB expression in liver cancer cell lines was achieved by RNA interfering technology to explore its biological function in HCC. RHEB expression was high in liver cancer tissues, with an increase of 2.00±0.19-fold compared with normal tissues and of 2.00±0.27-fold compared with adjacent non-cancer tissues. RHEB expression increased along with the clinical staging of HCC, and the overall survival and mortality of patients were closely correlated to RHEB levels, micro-vascular invasion, hepatitis B virus-DNA titer, tumor differentiation and pathological satellites (P<0.05). After knocking down RHEB in SMMC-7721 cells, the growth of liver cancer cells was significantly reduced. The majority of cells were blocked in S-phase, and their colony-forming and proliferating abilities significantly decreased (P<0.05). In vivo, upon downregulation of RHEB expression, the tumorigenic ability of HCC significantly decreased (P<0.05). These data suggest that RHEB expression is a significant prognostic factor and may be important in HCC cell growth. The present study highlights the importance of RHEB as a novel prognostic marker of HCC.
Collapse
Affiliation(s)
- Fuchen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Zeya Pan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Jinmin Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Junsheng Ni
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Chao Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Zhenguang Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Fangming Gu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Wei Dong
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Weiping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| |
Collapse
|
35
|
Chatterjee AD, Roy D, Guevara P, Pal R, Naryan M, Roychowdhury S, Das S. Arachidonic Acid Induces the Migration of MDA-MB-231 Cells by Activating Raft-associated Leukotriene B4 Receptors. CLINICAL CANCER DRUGS 2018; 5:28-41. [PMID: 30443489 PMCID: PMC6233886 DOI: 10.2174/2212697x05666180418145601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The migration of tumor cells is critical in spreading cancers through the lymphatic nodes and circulatory systems. Although arachidonic acid (AA) and its soluble metabolites have been shown to induce the migration of breast and colon cancer cells, the mechanism by which it induces such migration has not been fully understood. OBJECTIVE The effect of AA on migratory responses of the MDA-MB-231 cell line (a triple-negative breast cancer cell) was examined and compared with MCF-7 (estrogen-receptor positive) breast cancer cells to elucidate the mechanism of AA-induced migration. METHODS Migrations of breast cancer cells were examined with the help of wound-healing assays. AA-induced eicosanoid synthesis was monitored by RP-HPLC. Cellular localizations of lipoxygenase and lipid rafts were assessed by immunoblot and confocal microscopy. RESULTS AA treatment stimulated the synthesis of leukotriene B4 (LTB4) and HETE-8, but lowered the levels of prostaglandin E2 (PGE2), prostaglandin D2 (PGD2), and HETE-5 in MDA-MB-231 cells. Further analysis indicated that AA increased the expression of 5-lipoxygenase (5-LOX) in this cell line and inhibiting its expression by small molecule inhibitors lowered the production of LTB4 and reduced migration. In contrast, MCF-7 cells did not show any appreciable changes in eicosanoid synthesis, 5-LOX expression, or cellular migration. CONCLUSION Our results suggest that AA treatment activates the BLT1 receptor (present in membrane microdomains) and stimulates the synthesis of LTB4 production, which is likely to be associated with the migration of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Atasi De Chatterjee
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Debarshi Roy
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Priscilla Guevara
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Rituraj Pal
- Department of Chemistry, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Mahesh Naryan
- Department of Chemistry, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Sukla Roychowdhury
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Siddhartha Das
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| |
Collapse
|
36
|
Zanoaga O, Jurj A, Raduly L, Cojocneanu-Petric R, Fuentes-Mattei E, Wu O, Braicu C, Gherman CD, Berindan-Neagoe I. Implications of dietary ω-3 and ω-6 polyunsaturated fatty acids in breast cancer. Exp Ther Med 2017; 15:1167-1176. [PMID: 29434704 PMCID: PMC5776638 DOI: 10.3892/etm.2017.5515] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022] Open
Abstract
Breast cancer represents one of the most common forms of cancer in women worldwide, with an increase in the number of newly diagnosed patients in the last decade. The role of fatty acids, particularly of a diet rich in ω-3 and ω-6 polyunsaturated fatty acids (PUFAs), in breast cancer development is not fully understood and remains controversial due to their complex mechanism of action. However, a large number of animal models and cell culture studies have demonstrated that high levels of ω-3 PUFAs have an inhibitory role in the development and progression of breast cancer, compared to ω-6 PUFAs. The present review focused on recent studies regarding the correlation between dietary PUFAs and breast cancer development, and aimed to emphasize the main molecular mechanisms involved in the modification of cell membrane structure and function, modulation of signal transduction pathways, gene expression regulation, and antiangiogenic and antimetastatic effects. Furthermore, the anticancer role of ω-3 PUFAs through the modulation of microRNA expression levels was also reviewed.
Collapse
Affiliation(s)
- Oana Zanoaga
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.,Department of Physiopathology, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Roxana Cojocneanu-Petric
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Enrique Fuentes-Mattei
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Oscar Wu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Texas Tech University Honors College, McClellan Hall, Lubbock, TX 79409, USA
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Claudia Diana Gherman
- Surgical Clinic II Hospital, 400006 Cluj-Napoca, Romania.,Department of Surgery, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.,MEDFUTURE-Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, 400349 Cluj-Napoca, Romania.,Department of Functional Genomics, Proteomics and Experimental Pathology, Prof Dr Ion Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
| |
Collapse
|
37
|
Tsai CH, Shen YC, Chen HW, Liu KL, Chang JW, Chen PY, Lin CY, Yao HT, Li CC. Docosahexaenoic acid increases the expression of oxidative stress-induced growth inhibitor 1 through the PI3K/Akt/Nrf2 signaling pathway in breast cancer cells. Food Chem Toxicol 2017; 108:276-288. [DOI: 10.1016/j.fct.2017.08.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 10/19/2022]
|
38
|
Chen T, Li H. Fatty acid metabolism and prospects for targeted therapy of cancer. EUR J LIPID SCI TECH 2017. [DOI: 10.1002/ejlt.201600366] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Tingting Chen
- West China College of Basic and Forensic MedicineSichuan UniversityChengduP. R. China
| | - Hua Li
- West China College of Basic and Forensic MedicineSichuan UniversityChengduP. R. China
| |
Collapse
|
39
|
Dai YJ, Wang YY, Huang JY, Xia L, Shi XD, Xu J, Lu J, Su XB, Yang Y, Zhang WN, Wang PP, Wu SF, Huang T, Mi JQ, Han ZG, Chen Z, Chen SJ. Conditional knockin of Dnmt3a R878H initiates acute myeloid leukemia with mTOR pathway involvement. Proc Natl Acad Sci U S A 2017; 114:5237-5242. [PMID: 28461508 PMCID: PMC5441829 DOI: 10.1073/pnas.1703476114] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
DNMT3A is frequently mutated in acute myeloid leukemia (AML). To explore the features of human AML with the hotspot DNMT3A R882H mutation, we generated Dnmt3a R878H conditional knockin mice, which developed AML with enlarged Lin-Sca1+cKit+ cell compartments. The transcriptome and DNA methylation profiling of bulk leukemic cells and the single-cell RNA sequencing of leukemic stem/progenitor cells revealed significant changes in gene expression and epigenetic regulatory patterns that cause differentiation arrest and growth advantage. Consistent with leukemic cell accumulation in G2/M phase, CDK1 was up-regulated due to mTOR activation associated with DNA hypomethylation. Overexpressed CDK1-mediated EZH2 phosphorylation resulted in an abnormal trimethylation of H3K27 profile. The mTOR inhibitor rapamycin elicited a significant therapeutic response in Dnmt3aR878H/WT mice.
Collapse
Affiliation(s)
- Yu-Jun Dai
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yue-Ying Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Jin-Yan Huang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li Xia
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Dong Shi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jie Xu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Lu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xian-Bin Su
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying Yang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei-Na Zhang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Pan-Pan Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Song-Fang Wu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ting Huang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian-Qing Mi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ze-Guang Han
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhu Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Sai-Juan Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
40
|
Zuo Z, Syrovets T, Wu Y, Hafner S, Vernikouskaya I, Liu W, Ma G, Weil T, Simmet T, Rasche V. The CAM cancer xenograft as a model for initial evaluation of MR labelled compounds. Sci Rep 2017; 7:46690. [PMID: 28466861 PMCID: PMC5413881 DOI: 10.1038/srep46690] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 03/27/2017] [Indexed: 01/16/2023] Open
Abstract
Non-invasive assessment of the biodistribution is of great importance during the development of new pharmaceutical compounds. In this contribution, the applicability of in ovo MRI for monitoring the biodistribution of MR contrast agent-labelled compounds was investigated in mamaria carcinomas xentotransplanted on the chorioallantoic membrane (CAM) exemplarily for Gd-DOTA and cHSA-PEO (2000)16-Gd after systemic injection of the compounds into a chorioallantoic capillary vein. MRI was performed directly prior and 30 min, 3 h, 5 h, 20 h, and 40 h after injection of the compound. The biodistribution of injected compounds could be assessed by MRI in different organs of the chicken embryo as well as in xenotransplanted tumors at all time points. A clearly prolonged enhancement of the tumor substrate could be shown for cHSA-PEO (2000)16-Gd. In conclusion, high-resolution in ovo MR imaging can be used for assessment of the in vivo biodistribution of labelled compounds, thus enabling efficient non-invasive initial testing.
Collapse
Affiliation(s)
- Zhi Zuo
- Department of Internal Medicine II, University Hospital Ulm, Ulm, Germany.,Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China.,Core Facility Small Animal MRI, Medical Faculty, Ulm University, Ulm, Germany
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Yuzhou Wu
- Institute of Organic Chemistry III, Ulm University, Ulm, Germany
| | - Susanne Hafner
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Ina Vernikouskaya
- Department of Internal Medicine II, University Hospital Ulm, Ulm, Germany
| | - Weina Liu
- Institute of Organic Chemistry III, Ulm University, Ulm, Germany
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Tanja Weil
- Institute of Organic Chemistry III, Ulm University, Ulm, Germany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Volker Rasche
- Department of Internal Medicine II, University Hospital Ulm, Ulm, Germany.,Core Facility Small Animal MRI, Medical Faculty, Ulm University, Ulm, Germany
| |
Collapse
|
41
|
Wang H, Zhang H, Sun Q, Wang Y, Yang J, Yang J, Zhang T, Luo S, Wang L, Jiang Y, Zeng C, Cai D, Bai X. Intra-articular Delivery of Antago-miR-483-5p Inhibits Osteoarthritis by Modulating Matrilin 3 and Tissue Inhibitor of Metalloproteinase 2. Mol Ther 2017; 25:715-727. [PMID: 28139355 DOI: 10.1016/j.ymthe.2016.12.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/12/2016] [Accepted: 12/25/2016] [Indexed: 01/21/2023] Open
Abstract
MicroRNAs (miRNAs) are emerging as important regulators in osteoarthritis (OA) pathogenesis. In our study, a real-time PCR assay revealed that miR-483-5p was upregulated in articular cartilage from OA patients and experimental OA mice induced by destabilization of the medial meniscus compared to their controls. Overexpression of miR-483-5p by intra-articular injection of lentivirus LV3-miR-483-5p significantly enhanced the severity of experimental OA. Consequently, we synthesized antago-miR-483-5p to silence the endogenous miR-483-5p and delivered it intra-articularly, which revealed that antago-miR-483-5p delayed the progression of experimental OA. To investigate the functional mechanism of miR-483-5p in OA development, we generated doxycycline-inducible miR-483 transgenic (TG483) mice. TG483 mice exhibited significant acceleration and increased severity of OA, and age-related OA occurred with higher incidence and greater severity in TG483 mice compared with their controls. Furthermore, our results revealed miR-483-5p directly targeted to the cartilage matrix protein matrilin 3 (Matn3) and tissue inhibitor of metalloproteinase 2 (Timp2) to stimulate chondrocyte hypertrophy, extracellular matrix degradation, and cartilage angiogenesis, and it consequently initiated and accelerated the development of OA. In conclusion, our findings reveal an miRNA functional pathway important for OA development. Targeting of miR-483-5p by intra-articular injection of antago-miR-483-5p represents an approach that could prevent the onset of OA and delay its progression.
Collapse
Affiliation(s)
- Hua Wang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Tropical Diseases and Translational Medicine of The Ministry of Education, Hainan Medical College, Haikou 571199, China
| | - Haiyan Zhang
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Qiuyi Sun
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Yun Wang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Jun Yang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Jincheng Yang
- Department of Orthopedics, General Hospital of Guangzhou Military Command of PLA, Guangzhou 510010, China
| | - Tao Zhang
- Department of Orthopedics, General Hospital of Guangzhou Military Command of PLA, Guangzhou 510010, China
| | - Shenqiu Luo
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Liping Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chun Zeng
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| | - Daozhang Cai
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| | - Xiaochun Bai
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
42
|
Shi Y, Steppi A, Cao Y, Wang J, He MM, Li L, Zhang J. Integrative Comparison of mRNA Expression Patterns in Breast Cancers from Caucasian and Asian Americans with Implications for Precision Medicine. Cancer Res 2017; 77:423-433. [PMID: 28069798 PMCID: PMC5243181 DOI: 10.1158/0008-5472.can-16-1959] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/20/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022]
Abstract
Asian Americans (AS) have significantly lower incidence and mortality rates of breast cancer than Caucasian Americans (CA). Although this racial disparity has been documented, the underlying pathogenetic factors explaining it are obscure. We addressed this issue by an integrative genomics approach to compare mRNA expression between AS and CA cases of breast cancer. RNA-seq data from the Cancer Genome Atlas showed that mRNA expression revealed significant differences at gene and pathway levels. Increased susceptibility and severity in CA patients were likely the result of synergistic environmental and genetic risk factors, with arachidonic acid metabolism and PPAR signaling pathways implicated in linking environmental and genetic factors. An analysis that also added eQTL data from the Genotype-Tissue Expression Project and SNP data from the 1,000 Genomes Project identified several SNPs associated with differentially expressed genes. Overall, the associations we identified may enable a more focused study of genotypic differences that may help explain the disparity in breast cancer incidence and mortality rates in CA and AS populations and inform precision medicine. Cancer Res; 77(2); 423-33. ©2016 AACR.
Collapse
Affiliation(s)
- Yanxia Shi
- Department of medical oncology, Sun Yet-sen University cancer center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, State Key Laboratory of Oncology in South China, Guangdong, PR China
| | - Albert Steppi
- Department of statistics, Florida State University, Tallahassee, FL 32306
| | - Ye Cao
- Department of medical oncology, Sun Yet-sen University cancer center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, State Key Laboratory of Oncology in South China, Guangdong, PR China
| | - Jianan Wang
- No. 2 High school of East China Normal University, Shanghai, 201203, China
| | - Max M He
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI 54449, USA
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, WI 54449, USA
| | - Liren Li
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, WI 54449, USA
| | - Jinfeng Zhang
- Department of statistics, Florida State University, Tallahassee, FL 32306
| |
Collapse
|
43
|
Orlando UD, Castillo AF, Dattilo MA, Solano AR, Maloberti PM, Podesta EJ. Acyl-CoA synthetase-4, a new regulator of mTOR and a potential therapeutic target for enhanced estrogen receptor function in receptor-positive and -negative breast cancer. Oncotarget 2016; 6:42632-50. [PMID: 26536660 PMCID: PMC4767459 DOI: 10.18632/oncotarget.5822] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/09/2015] [Indexed: 01/15/2023] Open
Abstract
Although the role of acyl-CoA synthetase 4 (ACSL4) in mediating an aggressive phenotype is well accepted, there is little evidence as to the early steps through which ACSL4 increases tumor growth and progression. In this study, and by means of the stable transfection of MCF-7 cells with ACSL4 using the tetracycline Tet-Off system (MCF-7 Tet-Off/ACSL4), we identify the mTOR pathway as one of the main specific signatures of ACSL4 expression and demonstrate the partial involvement of the lipoxygenase pathway in the activation of mTOR. The specificity of ACSL4 action on mTOR signaling is also determined by doxycycline inhibition of ACSL4 expression in MCF-7 Tet-Off/ACSL4 cells, by the expression of ACSL4 in the non-aggressive T47D breast cancer cell line and by knocking down this enzyme expression in the MDA-MB-231 breast cancer cells, which constitutively express ACSL4. ACSL4 regulates components of the two complexes of the mTOR pathway (mTORC1/2), along with upstream regulators and substrates. We show that mTOR inhibitor rapamycin and ACSL4 inhibitor rosiglitazone can act in combination to inhibit cell growth. In addition, we demonstrate a synergistic effect on cell growth inhibition by the combination of rosiglitazone and tamoxifen, an estrogen receptor α (ERα) inhibitor. Remarkably, this synergistic effect is also evident in the triple negative MDA-MB-231 cells in vitro and in vivo. These results suggest that ACSL4 could be a target to restore tumor hormone dependence in tumors with poor prognosis for disease-free and overall survival, in which no effective specifically targeted therapy is readily available.
Collapse
Affiliation(s)
- Ulises D Orlando
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Ana F Castillo
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Melina A Dattilo
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Angela R Solano
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Paula M Maloberti
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Ernesto J Podesta
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| |
Collapse
|
44
|
Menopause-induced uterine epithelium atrophy results from arachidonic acid/prostaglandin E2 axis inhibition-mediated autophagic cell death. Sci Rep 2016; 6:31408. [PMID: 27506466 PMCID: PMC4979008 DOI: 10.1038/srep31408] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 07/21/2016] [Indexed: 02/05/2023] Open
Abstract
Women experience menopause later in life. Menopause is characterized by dramatically decreased circulating estrogen level secondary to loss of ovarian function and atrophic state of genital organs. However, the molecular mechanisms for this process are not fully understood. In this study, we aimed to investigate the potential molecular mechanisms that underlie menopause-induced uterine endometrial atrophy. Our data showed that autophagy was activated in the uterine epithelial cells of both ovariectomized rats and peri-menopausal females. Endoplasmic reticulum (ER) stress occurred even prior to autophagy induction. Integrated bioinformatics analysis revealed that ER stress induced downstream decreased release of arachidonic acid (AA) and downregulation of AA/prostaglandin E2 (PGE2) axis, which led to Akt/mTOR signaling pathway inactivation. Consequently, autophagosomes were recruited and LC3-dependent autophagy was induced in uterine epithelial cells. Treatment with exogenous E2, PGE2, salubrinal or RNAi-mediated silencing of key autophagy genes could effectively counteract estrogen depletion-induced autophagy. Collectively, autophagy is a critical regulator of the uterine epithelium that accounts for endometrial atrophy after menopause.
Collapse
|
45
|
Zhou Q, Jiang Y, Yin W, Wang Y, Lu J. Single-nucleotide polymorphism in microRNA-binding site of SULF1 target gene as a protective factor against the susceptibility to breast cancer: a case-control study. Onco Targets Ther 2016; 9:2749-57. [PMID: 27274271 PMCID: PMC4869662 DOI: 10.2147/ott.s102433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose Numerous clinical studies have suggested that chemopreventive drugs for breast cancer such as tamoxifen and exemestane can effectively reduce the incidence of estrogen receptor (ER)-positive breast cancer. However, it remains unclear how to identify those who are susceptible to ER-positive breast cancer. Accordingly, there is a great demand for a probe into the predisposing factors so as to provide precise chemoprevention. Recent evidence has indicated that ERα expression can be regulated by microRNAs (miRNAs), such as miR-206, in breast cancer. We assumed that single-nucleotide polymorphisms (SNPs) in the miR-206-binding sites of the target genes may be associated with breast cancer susceptibility with different ER statuses. Methods We genotyped the SNPs that reside in and around the miR-206-binding sites of two target genes – heparan sulfatase 1 (SULF1) and RPTOR-independent companion of mammalian target of rapamycin Complex 2 (RICTOR) – which were related to the progression or metastasis of breast cancer cells in 710 breast cancer patients and 294 controls by the matrix-assisted laser desorption ionization-time of flight mass spectrometry method. Modified odds ratios (ORs) with their 95% confidence intervals (CIs) were calculated by a multivariate logistic regression analysis to evaluate the potential association between the SNPs and breast cancer susceptibility. Results For rs3802278, which is located in the 3′-untranslated region (3′-UTR) of SULF1, the frequency of the AA genotype was less in breast cancer patients than that in the controls as compared to that of the GG + GA genotype not only for ER-positive breast cancer patients (adjusted OR =0.663, P=0.032) but also for hormone receptor-positive breast cancer patients (adjusted OR =0.610, P=0.018). Besides, the frequency of the AA genotype was less than that of the GG genotype between the ER-positive breast cancer patients and the controls (adjusted OR =0.791, P=0.038). For rs66916453, which is located in the 3′-UTR of RICTOR, no significant difference was observed between the case and the control group for the genotypes or alleles (P>0.05). Conclusion The SNPs in the miRNA-binding sites within the 3′-UTR of SULF1 may serve as protective factors against the susceptibility to breast cancer, especially to ER-positive breast cancer in the Chinese population. These SNPs are promising candidate biomarkers to predict the susceptibility of breast cancer and guide the administration of targeted preventive endocrine therapy.
Collapse
Affiliation(s)
- Qiong Zhou
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Department of Gynecology, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Yiwei Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Breast Cancer Center, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Wenjin Yin
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yaohui Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Breast Cancer Center, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jinsong Lu
- Breast Cancer Center, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| |
Collapse
|
46
|
Liu J, Xu M, Zhao Y, Ao C, Wu Y, Chen Z, Wang B, Bai X, Li M, Hu W. n-3 polyunsaturated fatty acids abrogate mTORC1/2 signaling and inhibit adrenocortical carcinoma growth in vitro and in vivo. Oncol Rep 2016; 35:3514-22. [PMID: 27035283 DOI: 10.3892/or.2016.4720] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/06/2016] [Indexed: 11/06/2022] Open
Abstract
n-3 polyunsaturated fatty acids (PUFAs) are essential for human health and have been reported to reduce the risk of cancer, inhibit the growth of various types of tumors both in vitro and in vivo, and affect adrenal function. However, their effects on adrenocortical carcinoma (ACC) are not known. In the present study, we demonstrated that docosahexenoic acid (DHA) inhibited ACC cell proliferation, colony formation and cell cycle progression, and promoted apoptosis. In addition, ectopic expression of fat-1, a desaturase that converts n-6 to n-3 PUFAs endogenously, also inhibited ACC cell proliferation. Moreover, supplementing n-3 PUFAs in the diet efficiently prevented ACC cell growth in xenograft models. Notably, implanted ACC cells were unable to grow in fat-1 transgenic severe combined immune deficiency mice. Further study revealed that exogenous and endogenous n-3 PUFAs efficiently suppressed both mTOR complex 1 (mTORC1) and mTORC2 signaling in ACC in vitro and in vivo. Taken together, our findings provide comprehensive preclinical evidence that n-3 PUFAs efficiently prevent ACC growth by inhibiting mTORC1/2, which may have important implications in the treatment of ACC.
Collapse
Affiliation(s)
- Jun Liu
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, P.R. China
| | - Meinian Xu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Yongbin Zhao
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, P.R. China
| | - Chunping Ao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Yukun Wu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Zhenguo Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Bangqi Wang
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, P.R. China
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Ming Li
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Weilie Hu
- Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, P.R. China
| |
Collapse
|
47
|
Kim E, Tunset HM, Cebulla J, Vettukattil R, Helgesen H, Feuerherm AJ, Engebråten O, Mælandsmo GM, Johansen B, Moestue SA. Anti-vascular effects of the cytosolic phospholipase A2 inhibitor AVX235 in a patient-derived basal-like breast cancer model. BMC Cancer 2016; 16:191. [PMID: 26951085 PMCID: PMC4782288 DOI: 10.1186/s12885-016-2225-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Group IVA cytosolic phospholipase A2 (cPLA2α) plays an important role in tumorigenesis and angiogenesis. It is overexpressed in basal-like breast cancer (BLBC), which is aggressive and usually triple-negative, making it unresponsive to current targeted therapies. Here, we evaluated the anti-angiogenic effects of a specific cPLA2α inhibitor, AVX235, in a patient-derived triple-negative BLBC model. METHODS Mice bearing orthotopic xenografts received i.p. injections of AVX235 or DMSO vehicle daily for 1 week and then every other day for up to 19 days. Six treated and six control mice were terminated after 2 days of treatment, and the tumors excised for high resolution magic angle spinning magnetic resonance spectroscopy (HR MAS MRS) and prostaglandin E2 (PGE2) enzyme immunoassay (EIA) analysis. A 1-week imaging study was performed on a separate cohort of mice. Longitudinal dynamic contrast enhanced (DCE)-MRI was performed before, after 4 days, and after 1 week of treatment. The mice were then perfused with a radiopaque vascular casting agent, and the tumors excised for micro-CT angiography. Subsequently, tumors were sectioned and stained with lectin and for Ki67 or α-smooth muscle actin to quantify endothelial cell proliferation and vessel maturity, respectively. Partial least squares discriminant analysis was performed on the multivariate HR MAS MRS data, and non-parametric univariate analyses using Mann-Whitney U tests (α = 0.05) were performed on all other data. RESULTS Glycerophosphocholine and PGE2 levels, measured by HR MAS MRS and EIA, respectively, were lower in treated tumors after 2 days of treatment. These molecular changes are expected downstream effects of cPLA2α inhibition and were followed by significant tumor growth inhibition after 8 days of treatment. DCE-MRI revealed that AVX235 treatment caused a decrease in tumor perfusion. Concordantly, micro-CT angiography showed that vessel volume fraction, density, and caliber were reduced in treated tumors. Moreover, histology showed decreased endothelial cell proliferation and fewer immature vessels in treated tumors. CONCLUSIONS These results demonstrate that cPLA2α inhibition with AVX235 resulted in decreased vascularization and perfusion and subsequent inhibition of tumor growth. Thus, cPLA2α inhibition may be a potential new therapeutic option for triple-negative basal-like breast cancer.
Collapse
Affiliation(s)
- Eugene Kim
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491, Trondheim, Norway.
| | - Hanna Maja Tunset
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491, Trondheim, Norway.
| | - Jana Cebulla
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491, Trondheim, Norway.
| | - Riyas Vettukattil
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491, Trondheim, Norway.
| | - Heidi Helgesen
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491, Trondheim, Norway.
| | - Astrid Jullumstrø Feuerherm
- Department of Biology, Norwegian University of Science and Technology, Realfagbygget, 7491, Trondheim, Norway.
| | - Olav Engebråten
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, P.O. Box 4953 Nydalen, Oslo, 0424, Norway.
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, P.O. Box 4953 Nydalen, Oslo, 0424, Norway.
| | - Berit Johansen
- Department of Biology, Norwegian University of Science and Technology, Realfagbygget, 7491, Trondheim, Norway.
| | - Siver Andreas Moestue
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491, Trondheim, Norway.
| |
Collapse
|
48
|
Munkhbayar S, Jang S, Cho AR, Choi SJ, Shin CY, Eun HC, Kim KH, Kwon O. Role of Arachidonic Acid in Promoting Hair Growth. Ann Dermatol 2016; 28:55-64. [PMID: 26848219 PMCID: PMC4737836 DOI: 10.5021/ad.2016.28.1.55] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/15/2015] [Accepted: 05/21/2015] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Arachidonic acid (AA) is an omega-6 polyunsaturated fatty acid present in all mammalian cell membranes, and involved in the regulation of many cellular processes, including cell survival, angiogenesis, and mitogenesis. The dermal papilla, composed of specialized fibroblasts located in the bulb of the hair follicle, contributes to the control of hair growth and the hair cycle. OBJECTIVE This study investigated the effect of AA on hair growth by using in vivo and in vitro models. METHODS The effect of AA on human dermal papilla cells (hDPCs) and hair shaft elongation was evaluated by MTT assay and hair follicle organ culture, respectively. The expression of various growth and survival factors in hDPCs were investigated by western blot or immunohistochemistry. The ability of AA to induce and prolong anagen phase in C57BL/6 mice was analyzed. RESULTS AA was found to enhance the viability of hDPCs and promote the expression of several factors responsible for hair growth, including fibroblast growth factor-7 (FGF-7) and FGF-10. Western blotting identified the role of AA in the phosphorylation of various transcription factors (ERK, CREB, and AKT) and increased expression of Bcl-2 in hDPCs. In addition, AA significantly promoted hair shaft elongation, with increased proliferation of matrix keratinocytes, during ex vivo hair follicle culture. It was also found to promote hair growth by induction and prolongation of anagen phase in telogen-stage C57BL/6 mice. CONCLUSION This study concludes that AA plays a role in promoting hair growth by increasing the expression of growth factors in hDPCs and enhancing follicle proliferation and survival.
Collapse
Affiliation(s)
- Semchin Munkhbayar
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
| | - Sunhyae Jang
- Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Korea
- Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - A-Ri Cho
- Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Korea
- Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Soon-Jin Choi
- Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Korea
- Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Chang Yup Shin
- Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Korea
- Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Hee Chul Eun
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyu Han Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
- Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Korea
- Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Ohsang Kwon
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
- Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Korea
- Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
49
|
Incorporating Biomarkers in Studies of Chemoprevention. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 882:69-94. [PMID: 26987531 DOI: 10.1007/978-3-319-22909-6_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite Food and Drug Administration approval of tamoxifen and raloxifene for breast cancer risk reduction and endorsement by multiple agencies, uptake of these drugs for primary prevention in the United States is only 4% for risk eligible women likely to benefit from their use. Side effects coupled with incomplete efficacy and lack of a survival advantage are the likely reasons. This disappointing uptake, after the considerable effort and expense of large Phase III cancer incidence trials required for approval, suggests that a new paradigm is required. Current prevention research is focused on (1) refining risk prediction, (2) exploring behavioral and natural product interventions, and (3) utilizing novel translational trial designs for efficacy. Risk biomarkers will play a central role in refining risk estimates from traditional models and selecting cohorts for prevention trials. Modifiable risk markers called surrogate endpoint or response biomarkers will continue to be used in Phase I and II prevention trials to determine optimal dose or exposure and likely effectiveness from an intervention. The majority of Phase II trials will continue to assess benign breast tissue for response and mechanism of action biomarkers. Co-trials are those in which human and animal cohorts receive the same effective dose and the same tissue biomarkers are assessed for modulation due to the intervention, but then additional animals are allowed to progress to cancer development. These collaborations linking biomarker modulation and cancer prevention may obviate the need for cancer incidence trials for non-prescription interventions.
Collapse
|
50
|
Kudryashova TV, Goncharov DA, Pena A, Ihida-Stansbury K, DeLisser H, Kawut SM, Goncharova EA. Profiling the role of mammalian target of rapamycin in the vascular smooth muscle metabolome in pulmonary arterial hypertension. Pulm Circ 2015; 5:667-80. [PMID: 26697174 DOI: 10.1086/683810] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Increased proliferation and resistance to apoptosis of pulmonary arterial vascular smooth muscle cells (PAVSMCs), coupled with metabolic reprogramming, are key components of pulmonary vascular remodeling, a major and currently irreversible pathophysiological feature of pulmonary arterial hypertension (PAH). We recently reported that activation of mammalian target of rapamycin (mTOR) plays a key role in increased energy generation and maintenance of the proliferative, apoptosis-resistant PAVSMC phenotype in human PAH, but the downstream effects of mTOR activation on PAH PAVSMC metabolism are not clear. Using liquid and gas chromatography-based mass spectrometry, we performed pilot metabolomic profiling of human microvascular PAVSMCs from idiopathic-PAH subjects before and after treatment with the selective adenosine triphosphate-competitive mTOR inhibitor PP242 and from nondiseased lungs. We have shown that PAH PAVSMCs have a distinct metabolomic signature of altered metabolites-components of fatty acid synthesis, deficiency of sugars, amino sugars, and nucleotide sugars-intermediates of protein and lipid glycosylation, and downregulation of key biochemicals involved in glutathione and nicotinamide adenine dinucleotide (NAD) metabolism. We also report that mTOR inhibition attenuated or reversed the majority of the PAH-specific abnormalities in lipogenesis, glycosylation, glutathione, and NAD metabolism without affecting altered polyunsaturated fatty acid metabolism. Collectively, our data demonstrate a critical role of mTOR in major PAH PAVSMC metabolic abnormalities and suggest the existence of de novo lipid synthesis in PAVSMCs in human PAH that may represent a new, important component of disease pathogenesis worthy of future investigation.
Collapse
Affiliation(s)
- Tatiana V Kudryashova
- Vascular Medicine Institute, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dmitry A Goncharov
- Vascular Medicine Institute, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andressa Pena
- Vascular Medicine Institute, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kaori Ihida-Stansbury
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Horace DeLisser
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven M Kawut
- Pulmonary Vascular Disease Program and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elena A Goncharova
- Vascular Medicine Institute, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|