1
|
Yu Q, Qu C, Liang J, Chen P, Zhang K, Zhang Y, Zhang Y, Li Z, Liu S, Yang Z, Sun H, Yang A. ESF1 and MIPEP proteins promote estrogen receptor-positive breast cancer proliferation and are associated with patient prognosis. Clin Proteomics 2024; 21:50. [PMID: 39004717 PMCID: PMC11247778 DOI: 10.1186/s12014-024-09502-8] [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: 02/21/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Estrogen receptor-positive (ER+) breast cancer accounts for two-thirds of all breast cancers, and its early and late recurrences still threaten patients' long-term survival and quality of life. Finding candidate tumor antigens and potential therapeutic targets is critical to addressing these unmet needs. METHOD The isobaric tags for relative and absolute quantitation (iTRAQ) proteomic analysis was employed to identify the differentially expressed proteins (DEPs) between ER + breast cancer and corresponding adjacent normal tissue. Candidate DEPs were screened by bioinformatic analyses, and their expression was confirmed by immunohistochemical (IHC) staining and western blot. A series of in vitro experiments, including wound healing assay, colony formation, and cell cycle assay, were performed to reveal the functions of selected DEPs. Additionally, their clinical significances were further analyzed. RESULT A total of 369 DEPs (fold change ≥ 2.0 or ≤ 0.66, P < 0.05) were discovered. Compared with normal tissue, 358 proteins were up-regulated and 11 proteins were down-regulated in ER + breast cancer. GO and KEGG enrichment analysis showed that DEPs were closely associated with RNA regulation and metabolic pathways. STRING analysis found ESF1 and MIPEP were the hub genes in breast cancer, whose increased expressions were verified by the IHC staining and western blot. Knocking down ESF1 and MIPEP inhibited colony formation and increased cell apoptosis. Besides, knocking down ESF1 inhibited wound healing but not MIPEP. In addition, ESF1 and MIPEP expression were negatively associated with patient prognosis. CONCLUSION The upregulation of ESF1 and MIPEP promoted ER + breast cancer proliferation, which might provide novel targets for the development of new therapies.
Collapse
Affiliation(s)
- Qing Yu
- Department of Clinical Laboratory, Shenzhen Kangning Hospital, Shenzhen, 518000, P. R. China
- Department of Clinical Laboratory, Maternal & Child Health Hospital of Foshan, Foshan, 528000, P.R. China
| | - Chunhua Qu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Jinliang Liang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Peiqi Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kaiying Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yanji Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yuening Zhang
- Zhongshan Medical College, Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Zherui Li
- The Third Clinical Medical School, Guangzhou Medical University, Guangzhou, 510180, P.R. China
| | - Shaojun Liu
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, P.R. China
| | - Zhaoshou Yang
- The First Affiliated Hospital, The First Clinical Medicine School of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, 510080, P.R. China.
| | - Hongyan Sun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Anli Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
2
|
Petridi S, Dubal D, Rikhy R, van den Ameele J. Mitochondrial respiration and dynamics of in vivo neural stem cells. Development 2022; 149:285126. [PMID: 36445292 PMCID: PMC10112913 DOI: 10.1242/dev.200870] [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: 12/02/2022]
Abstract
Neural stem cells (NSCs) in the developing and adult brain undergo many different transitions, tightly regulated by extrinsic and intrinsic factors. While the role of signalling pathways and transcription factors is well established, recent evidence has also highlighted mitochondria as central players in NSC behaviour and fate decisions. Many aspects of cellular metabolism and mitochondrial biology change during NSC transitions, interact with signalling pathways and affect the activity of chromatin-modifying enzymes. In this Spotlight, we explore recent in vivo findings, primarily from Drosophila and mammalian model systems, about the role that mitochondrial respiration and morphology play in NSC development and function.
Collapse
Affiliation(s)
- Stavroula Petridi
- Department of Clinical Neurosciences and MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Dnyanesh Dubal
- Department of Clinical Neurosciences and MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK.,Biology, Indian Institute of Science Education and Research, Homi Bhabha Road, Pashan, Pune 411008, India
| | - Richa Rikhy
- Biology, Indian Institute of Science Education and Research, Homi Bhabha Road, Pashan, Pune 411008, India
| | - Jelle van den Ameele
- Department of Clinical Neurosciences and MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| |
Collapse
|
3
|
Majumder S, Crabtree JS, Golde TE, Minter LM, Osborne BA, Miele L. Targeting Notch in oncology: the path forward. Nat Rev Drug Discov 2021; 20:125-144. [PMID: 33293690 DOI: 10.1038/s41573-020-00091-3] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
Notch signalling is involved in many aspects of cancer biology, including angiogenesis, tumour immunity and the maintenance of cancer stem-like cells. In addition, Notch can function as an oncogene and a tumour suppressor in different cancers and in different cell populations within the same tumour. Despite promising preclinical results and early-phase clinical trials, the goal of developing safe, effective, tumour-selective Notch-targeting agents for clinical use remains elusive. However, our continually improving understanding of Notch signalling in specific cancers, individual cancer cases and different cell populations, as well as crosstalk between pathways, is aiding the discovery and development of novel investigational Notch-targeted therapeutics.
Collapse
Affiliation(s)
- Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Judy S Crabtree
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Todd E Golde
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Lisa M Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Barbara A Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
4
|
Dergilev KV, Zubkova ЕS, Beloglazova IB, Menshikov МY, Parfyonova ЕV. Notch signal pathway - therapeutic target for regulation of reparative processes in the heart. TERAPEVT ARKH 2018; 90:112-121. [PMID: 30701843 DOI: 10.26442/00403660.2018.12.000014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Notch signaling pathway is a universal regulator of cell fate in embryogenesis and in maintaining the cell homeostasis of adult tissue. Through local cell-cell interactions, he controls neighboring cells behavior and determines their capacity for self-renewal, growth, survival, differentiation, and apoptosis. Recent studies have shown that the control of regenerative processes in the heart is also carried out with the participation of Notch system. At the heart of Notch regulates migration bone marrow progenitors and stimulates the proliferation of cardiomyocytes, cardiac progenitor cell activity, limits cardiomyocyte hypertrophy and fibrosis progression and stimulates angiogenesis. Notch signaling pathway may be regarded as a very promising target for the development of drugs for the stimulation of regeneration in the myocardium.
Collapse
Affiliation(s)
- K V Dergilev
- National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Е S Zubkova
- National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - I B Beloglazova
- National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - М Yu Menshikov
- National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Е V Parfyonova
- National Medical Research Center for Cardiology of the Ministry of Health of the Russian Federation, Moscow, Russia.,M.V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
5
|
Tomer D, Chippalkatti R, Mitra K, Rikhy R. ERK regulates mitochondrial membrane potential in fission deficient Drosophila follicle cells during differentiation. Dev Biol 2018; 434:48-62. [DOI: 10.1016/j.ydbio.2017.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/04/2017] [Accepted: 11/15/2017] [Indexed: 01/10/2023]
|
6
|
Xu J, Chi F, Guo T, Punj V, Lee WNP, French SW, Tsukamoto H. NOTCH reprograms mitochondrial metabolism for proinflammatory macrophage activation. J Clin Invest 2015; 125:1579-90. [PMID: 25798621 DOI: 10.1172/jci76468] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 02/09/2015] [Indexed: 12/20/2022] Open
Abstract
Metabolic reprogramming is implicated in macrophage activation, but the underlying mechanisms are poorly understood. Here, we demonstrate that the NOTCH1 pathway dictates activation of M1 phenotypes in isolated mouse hepatic macrophages (HMacs) and in a murine macrophage cell line by coupling transcriptional upregulation of M1 genes with metabolic upregulation of mitochondrial oxidative phosphorylation and ROS (mtROS) to augment induction of M1 genes. Enhanced mitochondrial glucose oxidation was achieved by increased recruitment of the NOTCH1 intracellular domain (NICD1) to nuclear and mitochondrial genes that encode respiratory chain components and by NOTCH-dependent induction of pyruvate dehydrogenase phosphatase 1 (Pdp1) expression, pyruvate dehydrogenase activity, and glucose flux to the TCA cycle. As such, inhibition of the NOTCH pathway or Pdp1 knockdown abrogated glucose oxidation, mtROS, and M1 gene expression. Conditional NOTCH1 deficiency in the myeloid lineage attenuated HMac M1 activation and inflammation in a murine model of alcoholic steatohepatitis and markedly reduced lethality following endotoxin-mediated fulminant hepatitis in mice. In vivo monocyte tracking further demonstrated the requirement of NOTCH1 for the migration of blood monocytes into the liver and subsequent M1 differentiation. Together, these results reveal that NOTCH1 promotes reprogramming of mitochondrial metabolism for M1 macrophage activation.
Collapse
MESH Headings
- Animals
- Cell Line
- Electron Transport/genetics
- Endotoxemia/complications
- Fatty Liver, Alcoholic/immunology
- Fatty Liver, Alcoholic/metabolism
- Fatty Liver, Alcoholic/pathology
- Feedback, Physiological
- Gene Expression Regulation
- Glucose/metabolism
- Inflammation/immunology
- Inflammation/metabolism
- Liver Failure, Acute/etiology
- Liver Failure, Acute/immunology
- Liver Failure, Acute/metabolism
- Liver Failure, Acute/pathology
- Macrophage Activation/genetics
- Macrophage Activation/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria/metabolism
- Myeloid Cells/metabolism
- Myeloid Cells/pathology
- Nitric Oxide/metabolism
- Oxidative Phosphorylation
- Protein Structure, Tertiary
- Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/antagonists & inhibitors
- Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/genetics
- Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/metabolism
- Pyruvate Dehydrogenase Complex/metabolism
- Reactive Oxygen Species/metabolism
- Receptor, Notch1/deficiency
- Receptor, Notch1/physiology
- Signal Transduction/physiology
- Transcription, Genetic
- Up-Regulation
Collapse
|
7
|
Whitaker HC, Shiong LL, Kay JD, Grönberg H, Warren AY, Seipel A, Wiklund F, Thomas B, Wiklund P, Miller JL, Menon S, Ramos-Montoya A, Vowler SL, Massie C, Egevad L, Neal DE. N-acetyl-L-aspartyl-L-glutamate peptidase-like 2 is overexpressed in cancer and promotes a pro-migratory and pro-metastatic phenotype. Oncogene 2014; 33:5274-87. [PMID: 24240687 DOI: 10.1038/onc.2013.464] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/27/2013] [Accepted: 09/16/2013] [Indexed: 02/02/2023]
Abstract
N-acetyl-L-aspartyl-L-glutamate peptidase-like 2 (NAALADL2) is a member of the glutamate carboxypeptidase II family, best characterized by prostate-specific membrane antigen (PSMA/NAALAD1). Using immunohistochemistry (IHC), we have shown overexpression of NAALADL2 in colon and prostate tumours when compared with benign tissue. In prostate cancer, NAALADL2 expression was associated with stage and Grade, as well as circulating mRNA levels of the NAALADL2 gene. Overexpression of NAALADL2 was shown to predict poor survival following radical prostatectomy. In contrast to PSMA/NAALAD1, NAALADL2 was localized at the basal cell surface where it promotes adhesion to extracellular matrix proteins. Using stable knockdown and overexpression cell lines, we have demonstrated NAALADL2-dependent changes in cell migration, invasion and colony-forming potential. Expression arrays of the knockdown and overexpression cell lines have identified nine genes that co-expressed with NAALADL2, which included membrane proteins and genes known to be androgen regulated, including the prostate cancer biomarkers AGR2 and SPON2. Androgen regulation was confirmed in a number of these genes, although NAALADL2 itself was not found to be androgen regulated. NAALADL2 was also found to regulate levels of Ser133 phosphorylated C-AMP-binding protein (CREB), a master regulator of a number of cellular processes involved in cancer development and progression. In combination, these data suggest that changes in expression of NAALADL2 can impact upon a number of pro-oncogenic pathways and processes, making it a useful biomarker for both diagnosis and prognosis.
Collapse
Affiliation(s)
- H C Whitaker
- 1] Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK [2] Cancer Research UK Biomarker Initiative, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - L L Shiong
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - J D Kay
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - H Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - A Y Warren
- 1] Department of Histopathology and ISH Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK [2] Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - A Seipel
- Department of Pathology, Karolinska Institute, Stockholm, Sweden
| | - F Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - B Thomas
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - P Wiklund
- Department of Pathology, Karolinska Institute, Stockholm, Sweden
| | - J L Miller
- 1] Department of Histopathology and ISH Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK [2] Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - S Menon
- Bioinformatics Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK
| | - A Ramos-Montoya
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - S L Vowler
- Bioinformatics Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK
| | - C Massie
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - L Egevad
- Department of Pathology, Karolinska Institute, Stockholm, Sweden
| | - D E Neal
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Yan HB, Lou ZZ, Li L, Brindley PJ, Zheng Y, Luo X, Hou J, Guo A, Jia WZ, Cai X. Genome-wide analysis of regulatory proteases sequences identified through bioinformatics data mining in Taenia solium. BMC Genomics 2014; 15:428. [PMID: 24899069 PMCID: PMC4070553 DOI: 10.1186/1471-2164-15-428] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 05/19/2014] [Indexed: 12/20/2022] Open
Abstract
Background Cysticercosis remains a major neglected tropical disease of humanity in many regions, especially in sub-Saharan Africa, Central America and elsewhere. Owing to the emerging drug resistance and the inability of current drugs to prevent re-infection, identification of novel vaccines and chemotherapeutic agents against Taenia solium and related helminth pathogens is a public health priority. The T. solium genome and the predicted proteome were reported recently, providing a wealth of information from which new interventional targets might be identified. In order to characterize and classify the entire repertoire of protease-encoding genes of T. solium, which act fundamental biological roles in all life processes, we analyzed the predicted proteins of this cestode through a combination of bioinformatics tools. Functional annotation was performed to yield insights into the signaling processes relevant to the complex developmental cycle of this tapeworm and to highlight a suite of the proteases as potential intervention targets. Results Within the genome of this helminth parasite, we identified 200 open reading frames encoding proteases from five clans, which correspond to 1.68% of the 11,902 protein-encoding genes predicted to be present in its genome. These proteases include calpains, cytosolic, mitochondrial signal peptidases, ubiquitylation related proteins, and others. Many not only show significant similarity to proteases in the Conserved Domain Database but have conserved active sites and catalytic domains. KEGG Automatic Annotation Server (KAAS) analysis indicated that ~60% of these proteases share strong sequence identities with proteins of the KEGG database, which are involved in human disease, metabolic pathways, genetic information processes, cellular processes, environmental information processes and organismal systems. Also, we identified signal peptides and transmembrane helices through comparative analysis with classes of important regulatory proteases. Phylogenetic analysis using Bayes approach provided support for inferring functional divergence among regulatory cysteine and serine proteases. Conclusion Numerous putative proteases were identified for the first time in T. solium, and important regulatory proteases have been predicted. This comprehensive analysis not only complements the growing knowledge base of proteolytic enzymes, but also provides a platform from which to expand knowledge of cestode proteases and to explore their biochemistry and potential as intervention targets. Electronic supplementary material The online version of this article (doi: 10.1186/1471-2164-15-428) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hong-Bin Yan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Key Laboratory of Veterinary Public Health of Agriculture Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu Province, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Zeidán-Chuliá F, de Oliveira BHN, Salmina AB, Casanova MF, Gelain DP, Noda M, Verkhratsky A, Moreira JCF. Altered expression of Alzheimer's disease-related genes in the cerebellum of autistic patients: a model for disrupted brain connectome and therapy. Cell Death Dis 2014; 5:e1250. [PMID: 24853428 PMCID: PMC4047885 DOI: 10.1038/cddis.2014.227] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/13/2014] [Accepted: 04/16/2014] [Indexed: 11/09/2022]
Abstract
Autism and Alzheimer's disease (AD) are, respectively, neurodevelopmental and degenerative diseases with an increasing epidemiological burden. The AD-associated amyloid-β precursor protein-α has been shown to be elevated in severe autism, leading to the 'anabolic hypothesis' of its etiology. Here we performed a focused microarray analysis of genes belonging to NOTCH and WNT signaling cascades, as well as genes related to AD and apoptosis pathways in cerebellar samples from autistic individuals, to provide further evidence for pathological relevance of these cascades for autism. By using the limma package from R and false discovery rate, we demonstrated that 31% (116 out of 374) of the genes belonging to these pathways displayed significant changes in expression (corrected P-values <0.05), with mitochondria-related genes being the most downregulated. We also found upregulation of GRIN1, the channel-forming subunit of NMDA glutamate receptors, and MAP3K1, known activator of the JNK and ERK pathways with anti-apoptotic effect. Expression of PSEN2 (presinilin 2) and APBB1 (or F65) were significantly lower when compared with control samples. Based on these results, we propose a model of NMDA glutamate receptor-mediated ERK activation of α-secretase activity and mitochondrial adaptation to apoptosis that may explain the early brain overgrowth and disruption of synaptic plasticity and connectome in autism. Finally, systems pharmacology analyses of the model that integrates all these genes together (NOWADA) highlighted magnesium (Mg(2+)) and rapamycin as most efficient drugs to target this network model in silico. Their potential therapeutic application, in the context of autism, is therefore discussed.
Collapse
Affiliation(s)
- F Zeidán-Chuliá
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - B-H N de Oliveira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - A B Salmina
- Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - M F Casanova
- Department of Psychiatry and Behavioral Sciences, University of Louisville, Louisville, KY, USA
| | - D P Gelain
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - M Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - A Verkhratsky
- 1] Faculty of Life Sciences, The University of Manchester, Manchester, UK [2] IKERBASQUE, Basque Foundation for Science, Bilbao, Spain [3] Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
| | - J C F Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
10
|
Basak NP, Roy A, Banerjee S. Alteration of mitochondrial proteome due to activation of Notch1 signaling pathway. J Biol Chem 2014; 289:7320-34. [PMID: 24474689 DOI: 10.1074/jbc.m113.519405] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Notch signaling pathway, a known regulator of cell fate decisions, proliferation, and apoptosis, has recently been implicated in the regulation of glycolysis, which affects tumor progression. However, the impact of Notch on other metabolic pathways remains to be elucidated. To gain more insights into the Notch signaling and its role in regulation of metabolism, we studied the mitochondrial proteome in Notch1-activated K562 cells using a comparative proteomics approach. The proteomic study led to the identification of 10 unique proteins that were altered due to Notch1 activation. Eight of these proteins belonged to mitochondria-localized metabolic pathways like oxidative phosphorylation, glutamine metabolism, Krebs cycle, and fatty acid oxidation. Validation of some of these findings showed that constitutive activation of Notch1 deregulated glutamine metabolism and Complex 1 of the respiratory chain. Furthermore, the deregulation of glutamine metabolism involved the canonical Notch signaling and its downstream effectors. The study also reports the effect of Notch signaling on mitochondrial function and status of high energy intermediates ATP, NADH, and NADPH. Thus our study shows the effect of Notch signaling on mitochondrial proteome, which in turn affects the functioning of key metabolic pathways, thereby connecting an important signaling pathway to the regulation of cellular metabolism.
Collapse
Affiliation(s)
- Nandini Pal Basak
- From the Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | | | | |
Collapse
|
11
|
Cama A, Verginelli F, Lotti LV, Napolitano F, Morgano A, D’Orazio A, Vacca M, Perconti S, Pepe F, Romani F, Vitullo F, di Lella F, Visone R, Mannelli M, Neumann HPH, Raiconi G, Paties C, Moschetta A, Tagliaferri R, Veronese A, Sanna M, Mariani-Costantini R. Integrative genetic, epigenetic and pathological analysis of paraganglioma reveals complex dysregulation of NOTCH signaling. Acta Neuropathol 2013; 126:575-94. [PMID: 23955600 PMCID: PMC3789891 DOI: 10.1007/s00401-013-1165-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 08/02/2013] [Indexed: 02/06/2023]
Abstract
Head and neck paragangliomas, rare neoplasms of the paraganglia composed of nests of neurosecretory and glial cells embedded in vascular stroma, provide a remarkable example of organoid tumor architecture. To identify genes and pathways commonly deregulated in head and neck paraganglioma, we integrated high-density genome-wide copy number variation (CNV) analysis with microRNA and immunomorphological studies. Gene-centric CNV analysis of 24 cases identified a list of 104 genes most significantly targeted by tumor-associated alterations. The "NOTCH signaling pathway" was the most significantly enriched term in the list (P = 0.002 after Bonferroni or Benjamini correction). Expression of the relevant NOTCH pathway proteins in sustentacular (glial), chief (neuroendocrine) and endothelial cells was confirmed by immunohistochemistry in 47 head and neck paraganglioma cases. There were no relationships between level and pattern of NOTCH1/JAG2 protein expression and germline mutation status in the SDH genes, implicated in paraganglioma predisposition, or the presence/absence of immunostaining for SDHB, a surrogate marker of SDH mutations. Interestingly, NOTCH upregulation was observed also in cases with no evidence of CNVs at NOTCH signaling genes, suggesting altered epigenetic modulation of this pathway. To address this issue we performed microarray-based microRNA expression analyses. Notably 5 microRNAs (miR-200a,b,c and miR-34b,c), including those most downregulated in the tumors, correlated to NOTCH signaling and directly targeted NOTCH1 in in vitro experiments using SH-SY5Y neuroblastoma cells. Furthermore, lentiviral transduction of miR-200s and miR-34s in patient-derived primary tympano-jugular paraganglioma cell cultures was associated with NOTCH1 downregulation and increased levels of markers of cell toxicity and cell death. Taken together, our results provide an integrated view of common molecular alterations associated with head and neck paraganglioma and reveal an essential role of NOTCH pathway deregulation in this tumor type.
Collapse
Affiliation(s)
- Alessandro Cama
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d’Annunzio University Foundation, Via Colle dell’Ara, 66100 Chieti, Italy
- Department of Pharmacy, G. d’Annunzio University, Via dei Vestini 1, 66100 Chieti, Italy
| | - Fabio Verginelli
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d’Annunzio University Foundation, Via Colle dell’Ara, 66100 Chieti, Italy
- Department of Pharmacy, G. d’Annunzio University, Via dei Vestini 1, 66100 Chieti, Italy
| | - Lavinia Vittoria Lotti
- Department of Experimental Medicine, University La Sapienza, Viale Regina Elena 324, 00161 Rome, Italy
| | - Francesco Napolitano
- NeuRoNe Lab, Department of Informatics, University of Salerno, Via Ponte Don Melillo, 84084 Fisciano, Salerno Italy
| | - Annalisa Morgano
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d’Annunzio University Foundation, Via Colle dell’Ara, 66100 Chieti, Italy
- Laboratory of Lipid Metabolism and Cancer, Department of Translational Pharmacology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti Italy
| | - Andria D’Orazio
- Laboratory of Lipid Metabolism and Cancer, Department of Translational Pharmacology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti Italy
| | - Michele Vacca
- Laboratory of Lipid Metabolism and Cancer, Department of Translational Pharmacology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti Italy
- IRCCS National Cancer Research Center Giovanni Paolo II, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Silvia Perconti
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d’Annunzio University Foundation, Via Colle dell’Ara, 66100 Chieti, Italy
- Department of Pharmacy, G. d’Annunzio University, Via dei Vestini 1, 66100 Chieti, Italy
| | - Felice Pepe
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d’Annunzio University Foundation, Via Colle dell’Ara, 66100 Chieti, Italy
- NeuRoNe Lab, Department of Informatics, University of Salerno, Via Ponte Don Melillo, 84084 Fisciano, Salerno Italy
| | - Federico Romani
- Department of Experimental Medicine, University La Sapienza, Viale Regina Elena 324, 00161 Rome, Italy
| | | | | | - Rosa Visone
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d’Annunzio University Foundation, Via Colle dell’Ara, 66100 Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University, Via dei Vestini 1, 66100 Chieti, Italy
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Hartmut P. H. Neumann
- Section of Preventive Medicine, Department of Nephrology, Albert-Ludwigs-University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
| | - Giancarlo Raiconi
- NeuRoNe Lab, Department of Informatics, University of Salerno, Via Ponte Don Melillo, 84084 Fisciano, Salerno Italy
| | - Carlo Paties
- Unit of Anatomic Pathology, Department of Clinical Pathology, Hospital G. da Saliceto, Via Giuseppe Taverna 49, 29100 Piacenza, Italy
| | - Antonio Moschetta
- Laboratory of Lipid Metabolism and Cancer, Department of Translational Pharmacology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti Italy
- IRCCS National Cancer Research Center Giovanni Paolo II, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Roberto Tagliaferri
- NeuRoNe Lab, Department of Informatics, University of Salerno, Via Ponte Don Melillo, 84084 Fisciano, Salerno Italy
| | - Angelo Veronese
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d’Annunzio University Foundation, Via Colle dell’Ara, 66100 Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University, Via dei Vestini 1, 66100 Chieti, Italy
| | - Mario Sanna
- Gruppo Otologico, Via Emmanueli 42, 29100 Piacenza, Italy
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University, Via dei Vestini 1, 66100 Chieti, Italy
| | - Renato Mariani-Costantini
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d’Annunzio University Foundation, Via Colle dell’Ara, 66100 Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University, Via dei Vestini 1, 66100 Chieti, Italy
| |
Collapse
|
12
|
Yu SD, Liu FY, Wang QR. Notch inhibitor: a promising carcinoma radiosensitizer. Asian Pac J Cancer Prev 2013; 13:5345-51. [PMID: 23317182 DOI: 10.7314/apjcp.2012.13.11.5345] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Radiotherapy is an important part of modern cancer management for many malignancies, and enhancing the radiosensitivity of tumor cells is critical for effective cancer therapies. The Notch signaling pathway plays a key role in regulation of numerous fundamental cellular processes. Further, there is accumulating evidence that dysregulated Notch activity is involved in the genesis of many human cancers. As such, Notch inhibitors are attractive therapeutic agents, although as for other anticancer agents, they exhibit significant and potential side effects. Thus, Notch inhibitors may be best used in combination with other agents or therapy. Herein, we describe evidence supporting the use of Notch inhibitors as novel and potent radiosensitizers in cancer therapy.
Collapse
Affiliation(s)
- Shu-Dong Yu
- Department of Otolaryngology, Qianfoshan Hospital Affiliated to Shandong University, Shandong, China.
| | | | | |
Collapse
|
13
|
Nolan MJ, Koga T, Walker L, McCarty R, Grybauskas A, Giovingo MC, Skuran K, Kuprys PV, Knepper PA. sCD44 internalization in human trabecular meshwork cells. Invest Ophthalmol Vis Sci 2013; 54:592-601. [PMID: 23287794 DOI: 10.1167/iovs.12-10627] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To determine whether soluble CD44 (sCD44), a likely biomarker of primary open-angle glaucoma (POAG), is internalized in cultured human trabecular meshwork (TM) cells and trafficked to mitochondria. METHODS In vitro, 32-kD sCD44 was isolated from human sera, biotinylated, and dephosphorylated. TM cells were incubated for 1 hour at 4°C with biotinylated albumin (b-albumin), biotin-labeled sCD44 (b-sCD44), or hypophosphorylated biotin-labeled sCD44 (-p b-sCD44) in the presence or absence of unlabeled sCD44, hyaluronic acid (HA), and a selected 10-mer HA binding peptide. The slides were warmed for 1 or 2 hours at 37°C, and 125 nM MitoTracker Red was added for the last 20 minutes of the incubation. The cells were washed, fixed, incubated with anti-biotin antibody and FITC-labeled goat anti-mouse antibody, and examined under a confocal microscope. RESULTS TM cell membranes were positive for b-sCD44 after 4°C incubation. When the temperature was raised to 37°C, b-sCD44 or -p b-sCD44 appeared in the cytoplasm. The internalization of b-sCD44 was blocked by excess unlabeled sCD44, HA, and a 10-mer HA-binding peptide. Double label experiments with b-sCD44 or -p b-sCD44 and MitoTracker Red indicated partial overlap. The percent co-localization of MitoTracker Red at 2 hours and FITC -p b-sCD44 was 17.4% (P < 0.001) and for FITC b-sCD44 was 11.7% (P < 0.001) compared with b-albumin. The influence of putative CD44 phosphorylation sites on mitochondrial trafficking was determined by TargetP 1.1. CONCLUSIONS sCD44 is internalized by TM cells and trafficked in part to mitochondria, which may be a factor in the toxicity of sCD44 in the POAG disease process.
Collapse
Affiliation(s)
- Michael J Nolan
- Department of Ophthalmology and Visual Sciences, University of Illinois-Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Gude N, Sussman M. Notch signaling and cardiac repair. J Mol Cell Cardiol 2012; 52:1226-32. [PMID: 22465038 DOI: 10.1016/j.yjmcc.2012.03.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/12/2012] [Accepted: 03/13/2012] [Indexed: 02/04/2023]
Abstract
Notch signaling is critical for proper heart development and recently has been reported to participate in adult cardiac repair. Notch resides at the cell surface as a single pass transmembrane receptor, transits through the cytoplasm following activation, and acts as a transcription factor upon entering the nucleus. This dynamic and widespread cellular distribution allows for potential interactions with many signaling and binding partners. Notch displays temporal as well as spatial versatility, acting as a strong developmental signal, controlling cell fate determination and lineage commitment, and playing a pivotal role in embryonic and adult stem cell proliferation and differentiation. This review serves as an update of recent literature addressing Notch signaling in the heart, with attention to findings from noncardiac research that provide clues for further interpretation of how the Notch pathway influences cardiac biology. Specific areas of focus include Notch signaling in adult myocardium following pathologic injury, the role of Notch in cardiac progenitor cells with respect to differentiation and cardiac repair, crosstalk between Notch and other cardiac signaling pathways, and emerging aspects of noncanonical Notch signaling in heart.
Collapse
Affiliation(s)
- Natalie Gude
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | | |
Collapse
|