1
|
Jian J, Yuan C, Hao H. Identifying key genes and functionally enriched pathways in acute myeloid leukemia by weighted gene co-expression network analysis. J Appl Genet 2024:10.1007/s13353-024-00881-0. [PMID: 38977582 DOI: 10.1007/s13353-024-00881-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 07/10/2024]
Abstract
Acute myeloid leukemia (AML) is characterized by the uncontrolled proliferation of myeloid leukemia cells in the bone marrow and other hematopoietic tissues and is highly heterogeneous. While with the progress of sequencing technology, understanding of the AML-related biomarkers is still incomplete. The purpose of this study is to identify potential biomarkers for prognosis of AML. Based on WGCNA analysis of gene mutation expression, methylation level distribution, mRNA expression, and AML-related genes in public databases were employed for investigating potential biomarkers for the prognosis of AML. This study screened a total of 6153 genes by analyzing various changes in 103 acute myeloid leukemia (AML) samples, including gene mutation expression, methylation level distribution, mRNA expression, and AML-related genes in public databases. Moreover, seven AML-related co-expression modules were mined by WGCNA analysis, and twelve biomarkers associated with the AML prognosis were identified from each top 10 genes of the seven co-expression modules. The AML samples were then classified into two subgroups, the prognosis of which is significantly different, based on the expression of these twelve genes. The differentially expressed 7 genes of two subgroups (HOXB-AS3, HOXB3, SLC9C2, CPNE8, MEG8, S1PR5, MIR196B) are mainly involved in glucose metabolism, glutathione biosynthesis, small G protein-mediated signal transduction, and the Rap1 signaling pathway. With the utilization of WGCNA mining, seven gene co-expression modules were identified from the TCGA database, and there are unreported genes that may be potential driver genes of AML and may be the direction to identify the possible molecular signatures to predict survival of AML patients and help guide experiments for potential clinical drug targets.
Collapse
Affiliation(s)
- Jimo Jian
- Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China
- Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Chenglu Yuan
- Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China
| | - Hongyuan Hao
- Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China.
| |
Collapse
|
2
|
Ramos-Molina B, Rossell J, Pérez-Montes de Oca A, Pardina E, Genua I, Rojo-López MI, Julián MT, Alonso N, Julve J, Mauricio D. Therapeutic implications for sphingolipid metabolism in metabolic dysfunction-associated steatohepatitis. Front Endocrinol (Lausanne) 2024; 15:1400961. [PMID: 38962680 PMCID: PMC11220194 DOI: 10.3389/fendo.2024.1400961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), a leading cause of chronic liver disease, has increased worldwide along with the epidemics of obesity and related dysmetabolic conditions characterized by impaired glucose metabolism and insulin signaling, such as type 2 diabetes mellitus (T2D). MASLD can be defined as an excessive accumulation of lipid droplets in hepatocytes that occurs when the hepatic lipid metabolism is totally surpassed. This metabolic lipid inflexibility constitutes a central node in the pathogenesis of MASLD and is frequently linked to the overproduction of lipotoxic species, increased cellular stress, and mitochondrial dysfunction. A compelling body of evidence suggests that the accumulation of lipid species derived from sphingolipid metabolism, such as ceramides, contributes significantly to the structural and functional tissue damage observed in more severe grades of MASLD by triggering inflammatory and fibrogenic mechanisms. In this context, MASLD can further progress to metabolic dysfunction-associated steatohepatitis (MASH), which represents the advanced form of MASLD, and hepatic fibrosis. In this review, we discuss the role of sphingolipid species as drivers of MASH and the mechanisms involved in the disease. In addition, given the absence of approved therapies and the limited options for treating MASH, we discuss the feasibility of therapeutic strategies to protect against MASH and other severe manifestations by modulating sphingolipid metabolism.
Collapse
Affiliation(s)
- Bruno Ramos-Molina
- Group of Obesity, Diabetes & Metabolism, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Joana Rossell
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Eva Pardina
- Department de Biochemistry & Molecular Biology, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Idoia Genua
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marina I. Rojo-López
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
| | - María Teresa Julián
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Núria Alonso
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Josep Julve
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Didac Mauricio
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Faculty of Medicine, University of Vic/Central University of Catalonia (UVIC/UCC), Vic, Spain
| |
Collapse
|
3
|
de los Ríos C, Viejo L, Carretero VJ, Juárez NH, Cruz-Martins N, Hernández-Guijo JM. Promising Molecular Targets in Pharmacological Therapy for Neuronal Damage in Brain Injury. Antioxidants (Basel) 2023; 12:118. [PMID: 36670980 PMCID: PMC9854812 DOI: 10.3390/antiox12010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
The complex etiopathogenesis of brain injury associated with neurodegeneration has sparked a lot of studies in the last century. These clinical situations are incurable, and the currently available therapies merely act on symptoms or slow down the course of the diseases. Effective methods are being sought with an intent to modify the disease, directly acting on the properly studied targets, as well as to contribute to the development of effective therapeutic strategies, opening the possibility of refocusing on drug development for disease management. In this sense, this review discusses the available evidence for mitochondrial dysfunction induced by Ca2+ miscommunication in neurons, as well as how targeting phosphorylation events may be used to modulate protein phosphatase 2A (PP2A) activity in the treatment of neuronal damage. Ca2+ tends to be the catalyst for mitochondrial dysfunction, contributing to the synaptic deficiency seen in brain injury. Additionally, emerging data have shown that PP2A-activating drugs (PADs) suppress inflammatory responses by inhibiting different signaling pathways, indicating that PADs may be beneficial for the management of neuronal damage. In addition, a few bioactive compounds have also triggered the activation of PP2A-targeted drugs for this treatment, and clinical studies will help in the authentication of these compounds. If the safety profiles of PADs are proven to be satisfactory, there is a case to be made for starting clinical studies in the setting of neurological diseases as quickly as possible.
Collapse
Affiliation(s)
- Cristóbal de los Ríos
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, University Rey Juan Carlos, Avda. Atenas s/n, 28922 Alcorcón, Spain
| | - Lucía Viejo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Victoria Jiménez Carretero
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natalia Hernández Juárez
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natália Cruz-Martins
- Faculty of Medicine, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Advanced Training in Health Sciences and Technologies, Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal
| | - Jesús M. Hernández-Guijo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Ramón y Cajal Institute for Health Research, IRYCIS, Hospital Ramón y Cajal, Ctra. de Colmenar Viejo, Km. 9,100, 28029 Madrid, Spain
| |
Collapse
|
4
|
Agapito G, Milano M, Cannataro M. A statistical network pre-processing method to improve relevance and significance of gene lists in microarray gene expression studies. BMC Bioinformatics 2022; 23:393. [PMID: 36167506 PMCID: PMC9516794 DOI: 10.1186/s12859-022-04936-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Microarrays can perform large scale studies of differential expressed gene (DEGs) and even single nucleotide polymorphisms (SNPs), thereby screening thousands of genes for single experiment simultaneously. However, DEGs and SNPs are still just as enigmatic as the first sequence of the genome. Because they are independent from the affected biological context. Pathway enrichment analysis (PEA) can overcome this obstacle by linking both DEGs and SNPs to the affected biological pathways and consequently to the underlying biological functions and processes. RESULTS To improve the enrichment analysis results, we present a new statistical network pre-processing method by mapping DEGs and SNPs on a biological network that can improve the relevance and significance of the DEGs or SNPs of interest to incorporate pathway topology information into the PEA. The proposed methodology improves the statistical significance of the PEA analysis in terms of computed p value for each enriched pathways and limit the number of enriched pathways. This helps reduce the number of relevant biological pathways with respect to a non-specific list of genes. CONCLUSION The proposed method provides two-fold enhancements. Network analysis reveals fewer DEGs, by selecting only relevant DEGs and the detected DEGs improve the enriched pathways' statistical significance, rather than simply using a general list of genes.
Collapse
Affiliation(s)
- Giuseppe Agapito
- Department of Law, Economics and Sociology Sciences, University Magna Græcia, 88100 Catanzaro, Italy
- Data Analytics Research Center, University Magna Græcia, 88100 Catanzaro, Italy
| | - Marianna Milano
- Data Analytics Research Center, University Magna Græcia, 88100 Catanzaro, Italy
- Department of Medical and Surgical Sciences, University Magna Græcia, 88100 Catanzaro, Italy
| | - Mario Cannataro
- Data Analytics Research Center, University Magna Græcia, 88100 Catanzaro, Italy
- Department of Medical and Surgical Sciences, University Magna Græcia, 88100 Catanzaro, Italy
| |
Collapse
|
5
|
Scarpa M, Singh P, Bailey CM, Lee JK, Kapoor S, Lapidus RG, Niyongere S, Sangodkar J, Wang Y, Perrotti D, Narla G, Baer MR. PP2A-activating Drugs Enhance FLT3 Inhibitor Efficacy through AKT Inhibition-Dependent GSK-3β-Mediated c-Myc and Pim-1 Proteasomal Degradation. Mol Cancer Ther 2021; 20:676-690. [PMID: 33568357 PMCID: PMC8027945 DOI: 10.1158/1535-7163.mct-20-0663] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/26/2020] [Indexed: 11/16/2022]
Abstract
Fms-like tyrosine-like kinase 3 internal tandem duplication (FLT3-ITD) is present in acute myeloid leukemia (AML) in 30% of patients and is associated with short disease-free survival. FLT3 inhibitor efficacy is limited and transient but may be enhanced by multitargeting of FLT3-ITD signaling pathways. FLT3-ITD drives both STAT5-dependent transcription of oncogenic Pim-1 kinase and inactivation of the tumor-suppressor protein phosphatase 2A (PP2A), and FLT3-ITD, Pim-1, and PP2A all regulate the c-Myc oncogene. We studied mechanisms of action of cotreatment of FLT3-ITD-expressing cells with FLT3 inhibitors and PP2A-activating drugs (PADs), which are in development. PADs, including FTY720 and DT-061, enhanced FLT3 inhibitor growth suppression and apoptosis induction in FLT3-ITD-expressing cell lines and primary AML cells in vitro and MV4-11 growth suppression in vivo PAD and FLT3 inhibitor cotreatment independently downregulated c-Myc and Pim-1 protein through enhanced proteasomal degradation. c-Myc and Pim-1 downregulation was preceded by AKT inactivation, did not occur in cells expressing myristoylated (constitutively active) AKT1, and could be induced by AKT inhibition. AKT inactivation resulted in activation of GSK-3β, and GSK-3β inhibition blocked downregulation of both c-Myc and Pim-1 by PAD and FLT3 inhibitor cotreatment. GSK-3β activation increased c-Myc proteasomal degradation through c-Myc phosphorylation on T58; infection with c-Myc with T58A substitution, preventing phosphorylation, blocked downregulation of c-Myc by PAD and FLT3 inhibitor cotreatment. GSK-3β also phosphorylated Pim-1L/Pim-1S on S95/S4. Thus, PADs enhance efficacy of FLT3 inhibitors in FLT3-ITD-expressing cells through a novel mechanism involving AKT inhibition-dependent GSK-3β-mediated increased c-Myc and Pim-1 proteasomal degradation.
Collapse
Affiliation(s)
- Mario Scarpa
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Medicine
| | - Prerna Singh
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
| | - Christopher M Bailey
- Department of Surgery and
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jonelle K Lee
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
| | - Shivani Kapoor
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
| | - Rena G Lapidus
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Medicine
| | - Sandrine Niyongere
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Medicine
| | - Jaya Sangodkar
- Division of Genetic Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Yin Wang
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Surgery and
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Danilo Perrotti
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Medicine
| | - Goutham Narla
- Division of Genetic Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Maria R Baer
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center,
- Department of Medicine
- Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
6
|
Gilad Y, Eliaz Y, Yu Y, Dean AM, Han SJ, Qin L, O’Malley BW, Lonard DM. A genome-scale CRISPR Cas9 dropout screen identifies synthetically lethal targets in SRC-3 inhibited cancer cells. Commun Biol 2021; 4:399. [PMID: 33767353 PMCID: PMC7994904 DOI: 10.1038/s42003-021-01929-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/24/2021] [Indexed: 02/01/2023] Open
Abstract
Steroid receptor coactivator 3 (SRC-3/NCoA3/AIB1), is a key regulator of gene transcription and it plays a central role in breast cancer (BC) tumorigenesis, making it a potential therapeutic target. Beyond its function as an important regulator of estrogen receptor transcriptional activity, SRC-3 also functions as a coactivator for a wide range of other transcription factors, suggesting SRC-3 inhibition can be beneficial in hormone-independent cancers as well. The recent discovery of a potent SRC-3 small molecule inhibitor, SI-2, enabled the further development of additional related compounds. SI-12 is an improved version of SI-2 that like SI-2 has anti-proliferative activity in various cancer types, including BC. Here, we sought to identify gene targets, that when inhibited in the presence of SI-12, would lead to enhanced BC cell cytotoxicity. We performed a genome-scale CRISPR-Cas9 screen in MCF-7 BC cells under conditions of pharmacological pressure with SI-12. A parallel screen was performed with an ER inhibitor, fulvestrant, to shed light on both common and distinct activities between SRC-3 and ERα inhibition. Bearing in mind the key role of SRC-3 in tumorigenesis of other types of cancer, we extended our study by validating potential hits identified from the MCF-7 screen in other cancer cell lines.
Collapse
Affiliation(s)
- Yosi Gilad
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Yossi Eliaz
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX USA
| | - Yang Yu
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Adam M. Dean
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - San Jung Han
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Li Qin
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Bert W. O’Malley
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - David M. Lonard
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
7
|
Costa B, Amorim I, Gärtner F, Vale N. Understanding Breast cancer: from conventional therapies to repurposed drugs. Eur J Pharm Sci 2020; 151:105401. [PMID: 32504806 DOI: 10.1016/j.ejps.2020.105401] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 04/22/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
Breast cancer is the most common cancer among women and is considered a developed country disease. Moreover, is a heterogenous disease, existing different types and stages of breast cancer development, therefore, better understanding of cancer biology, helps to improve the development of therapies. The conventional treatments accessible after diagnosis, have the main goal of controlling the disease, by improving survival. In more advance stages the aim is to prolong life and symptom palliation care. Surgery, radiation therapy and chemotherapy are the main options available, which must be adapted to each person individually. However, patients are developing resistance to the conventional therapies. This resistance is due to alterations in important regulatory pathways such as PI3K/AKt/mTOR, this pathway contributes to trastuzumab resistance, a reference drug to treat breast cancer. Therefore, is proposed the repurposing of drugs, instead of developing drugs de novo, for example, to seek new medical treatments within the drugs available, to be used in breast cancer treatment. Providing safe and tolerable treatments to patients, and new insights to efficacy and efficiency of breast cancer treatments. The economic and social burden of cancer is enormous so it must be taken measures to relieve this burden and to ensure continued access to therapies to all patients. In this review we focus on how conventional therapies against breast cancer are leading to resistance, by reviewing those mechanisms and discussing the efficacy of repurposed drugs to fight breast cancer.
Collapse
Affiliation(s)
- Bárbara Costa
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo 228, 4050-313 Porto, Portugal
| | - Irina Amorim
- Department of Molecular Pathology and Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo 228, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal; i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen, 4200-135 Porto, Portugal
| | - Fátima Gärtner
- Department of Molecular Pathology and Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo 228, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal; i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen, 4200-135 Porto, Portugal
| | - Nuno Vale
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo 228, 4050-313 Porto, Portugal; Department of Molecular Pathology and Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo 228, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal; i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen, 4200-135 Porto, Portugal.
| |
Collapse
|
8
|
Vicente C, Arriazu E, Martínez-Balsalobre E, Peris I, Marcotegui N, García-Ramírez P, Pippa R, Rabal O, Oyarzábal J, Guruceaga E, Prósper F, Mateos MC, Cayuela ML, Odero MD. A novel FTY720 analogue targets SET-PP2A interaction and inhibits growth of acute myeloid leukemia cells without inducing cardiac toxicity. Cancer Lett 2020; 468:1-13. [DOI: 10.1016/j.canlet.2019.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 10/25/2022]
|
9
|
Huang X, Lin H, Huang F, Xie Y, Wong KH, Chen X, Wu D, Lu A, Yang Z. Targeting Approaches of Nanomedicines in Acute Myeloid Leukemia. Dose Response 2019; 17:1559325819887048. [PMID: 31853234 PMCID: PMC6906351 DOI: 10.1177/1559325819887048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/10/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy, which is commonly
associated with high incidence and mortality among adult patients. The standard
induction regimen for AML has been substantially unchanged over the past 40
years, for which novel nanomedicines have represented a promising strategy in
AML therapies. Despite developments of multiple nanoparticles formulated with
drugs or genes, less there is not much information available about approaches in
AML is available. This review presents an overview of nanomedicines currently
being evaluated in AML. First, it briefly summarized conventional chemotherapies
in use. Second, nanomedicines presently ongoing in clinical trials or
preclinical researches were classified and described, with illustrative examples
from recent literatures. Finally, limitations and potential safety issues
concerns in clinical translation of AML treatment were discussed as well.
Collapse
Affiliation(s)
- Xiao Huang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hai Lin
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Feng Huang
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuning Xie
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ka Hong Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Dongyue Wu
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
10
|
FTY720 induces non-canonical phosphatidylserine externalization and cell death in acute myeloid leukemia. Cell Death Dis 2019; 10:847. [PMID: 31699964 PMCID: PMC6838108 DOI: 10.1038/s41419-019-2080-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/15/2019] [Accepted: 10/21/2019] [Indexed: 01/01/2023]
Abstract
FTY720 (fingolimod) is a FDA-approved sphingosine analog that is phosphorylated in vivo to modulate sphingosine-1-phosphate receptor (S1PR) signaling for immunosuppression in patients with refractory multiple sclerosis. FTY720 also exhibits promising anticancer efficacy in several preclinical models. While FTY720-induced cytotoxicity is not due to S1PR signaling, the mechanism remains unclear and is reported to occur through various cell death pathways. Here, we performed a systematic, mechanistic study of FTY720-induced cell death in acute myeloid leukemia (AML). We found that FTY720 induced cell death in a panel of genetically diverse AML cell lines that was accompanied by rapid phosphatidylserine (PS) externalization. Importantly, FTY720-induced PS exposure was not due to any direct effects on plasma membrane integrity and was independent of canonical signaling by regulated cell death pathways known to activate lipid flip-flop, including caspase-dependent apoptosis/pyroptosis, necroptosis, ferroptosis, and reactive oxygen species-mediated cell death. Notably, PS exposure required cellular vacuolization induced by defects in endocytic trafficking and was suppressed by the inhibition of PP2A and shedding of Annexin V-positive subcellular particles. Collectively, our studies reveal a non-canonical pathway underlying PS externalization and cell death in AML to provide mechanistic insight into the antitumor properties of FTY720.
Collapse
|
11
|
Clark AR, Ohlmeyer M. Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther 2019; 201:181-201. [PMID: 31158394 PMCID: PMC6700395 DOI: 10.1016/j.pharmthera.2019.05.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric enzyme that catalyzes the selective removal of phosphate groups from protein serine and threonine residues. Emerging evidence suggests that it functions as a tumor suppressor by constraining phosphorylation-dependent signalling pathways that regulate cellular transformation and metastasis. Therefore, PP2A-activating drugs (PADs) are being actively sought and investigated as potential novel anti-cancer treatments. Here we explore the concept that PP2A also constrains inflammatory responses through its inhibitory effects on various signalling pathways, suggesting that PADs may be effective in the treatment of inflammation-mediated pathologies.
Collapse
Affiliation(s)
- Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
| | | |
Collapse
|
12
|
Briot T, Roger E, Thépot S, Lagarce F. Advances in treatment formulations for acute myeloid leukemia. Drug Discov Today 2018; 23:1936-1949. [DOI: 10.1016/j.drudis.2018.05.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/25/2018] [Accepted: 05/29/2018] [Indexed: 10/24/2022]
|
13
|
Gonzalez D, Arribas RL, Viejo L, Lajarin-Cuesta R, de los Rios C. Substituent effect of N-benzylated gramine derivatives that prevent the PP2A inhibition and dissipate the neuronal Ca2+ overload, as a multitarget strategy for the treatment of Alzheimer’s disease. Bioorg Med Chem 2018; 26:2551-2560. [DOI: 10.1016/j.bmc.2018.04.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/06/2018] [Accepted: 04/07/2018] [Indexed: 11/28/2022]
|
14
|
Smith AM, Dun MD, Lee EM, Harrison C, Kahl R, Flanagan H, Panicker N, Mashkani B, Don AS, Morris J, Toop H, Lock RB, Powell JA, Thomas D, Guthridge MA, Moore A, Ashman LK, Skelding KA, Enjeti A, Verrills NM. Activation of protein phosphatase 2A in FLT3+ acute myeloid leukemia cells enhances the cytotoxicity of FLT3 tyrosine kinase inhibitors. Oncotarget 2018; 7:47465-47478. [PMID: 27329844 PMCID: PMC5216954 DOI: 10.18632/oncotarget.10167] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/06/2016] [Indexed: 11/25/2022] Open
Abstract
Constitutive activation of the receptor tyrosine kinase Fms-like tyrosine kinase 3 (FLT3), via co-expression of its ligand or by genetic mutation, is common in acute myeloid leukemia (AML). In this study we show that FLT3 activation inhibits the activity of the tumor suppressor, protein phosphatase 2A (PP2A). Using BaF3 cells transduced with wildtype or mutant FLT3, we show that FLT3-induced PP2A inhibition sensitizes cells to the pharmacological PP2A activators, FTY720 and AAL(S). FTY720 and AAL(S) induced cell death and inhibited colony formation of FLT3 activated cells. Furthermore, PP2A activators reduced the phosphorylation of ERK and AKT, downstream targets shared by both FLT3 and PP2A, in FLT3/ITD+ BaF3 and MV4-11 cell lines. PP2A activity was lower in primary human bone marrow derived AML blasts compared to normal bone marrow, with blasts from FLT3-ITD patients displaying lower PP2A activity than WT-FLT3 blasts. Reduced PP2A activity was associated with hyperphosphorylation of the PP2A catalytic subunit, and reduced expression of PP2A structural and regulatory subunits. AML patient blasts were also sensitive to cell death induced by FTY720 and AAL(S), but these compounds had minimal effect on normal CD34+ bone marrow derived monocytes. Finally, PP2A activating compounds displayed synergistic effects when used in combination with tyrosine kinase inhibitors in FLT3-ITD+ cells. A combination of Sorafenib and FTY720 was also synergistic in the presence of a protective stromal microenvironment. Thus combining a PP2A activating compound and a FLT3 inhibitor may be a novel therapeutic approach for treating AML.
Collapse
Affiliation(s)
- Amanda M Smith
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Current address: The University of Queensland Diamantina Institute, Woolloongabba, Queensland, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Erwin M Lee
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW, Sydney, New South Wales, Australia
| | - Celeste Harrison
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Richard Kahl
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Hayley Flanagan
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Nikita Panicker
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Baratali Mashkani
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Current address: Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Anthony S Don
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Jonathan Morris
- School of Chemistry, University of New South Wales, Sydney, New South Wales, Australia
| | - Hamish Toop
- School of Chemistry, University of New South Wales, Sydney, New South Wales, Australia
| | - Richard B Lock
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW, Sydney, New South Wales, Australia
| | - Jason A Powell
- Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | - Daniel Thomas
- Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia.,Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Mark A Guthridge
- Department Clinical Haematology, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Andrew Moore
- Translational Research Institute, The University of Queensland Diamantina Institute, Woolloongabba, Queensland, Australia
| | - Leonie K Ashman
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Kathryn A Skelding
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Anoop Enjeti
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Calvary Mater Hospital, Newcastle, New South Wales, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| |
Collapse
|
15
|
Wang H, Zhang Y, Li H, Zeng W, Qiao M. Shuyu capsules relieve liver- qi depression by regulating ERK-CREB-BDNF signal pathway in central nervous system of rat. Exp Ther Med 2017; 14:4831-4838. [PMID: 29201187 PMCID: PMC5704346 DOI: 10.3892/etm.2017.5125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/11/2017] [Indexed: 01/26/2023] Open
Abstract
The purpose of this study was to investigate the possible therapeutic mechanism of Shuyu capsules in liver-qi depression. Liver-qi depression rats were prepared based on chronic unpredictable mild stress (CUMS) and delayed constraint. Rats were gavaged with Shuyu capsule, fluoxetine, Radix Bupleuri and Radix Paeoniae Alba to constrct rat models. Body weight test, sucrose preference test and open-field test were applied to test rat models. Western blot analysis and quantitative real-time PCR was applied to determine the relative expression of extracellular signal-regulated protein kinase (ERK), cyclic AMP response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF) in hippocampus and frontal lobe tissues. ELISA was used to detect the content of BDNF in serum. Body weight, sugar intake and total distance were significantly decreased in depression group compared with control. The four drugs significantly increased levels of these factors. Compared with control group, ERK, CREB and BDNF expression were significantly decreased in depression group in both hippocampus and frontal lobe tissues at both mRNA and protein level. Shuyu capsule and fluoxetine group showed a significant increase in the expression of ERK, CREB and BDNF at mRNA, p-ERK and p-BDNF at protein level. Compared with Radix Paeoniae Alba, Radix Bupleuri were better in the rescue of ERK, CREB and BDNF expression. In conclusion, the pathogenesis of liver-qi depression associated with lower expression of ERK, CREB and BDNF in hippocampus and frontal. Shuyu capsule and main constitution alleviated the depressive-like behaviors and reversed the disruptions of the p-ERK, p-CREB and BDNF in stressed rats.
Collapse
Affiliation(s)
- Hongyan Wang
- Department of Traditional Chinese Medicine, Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| | - Yingquan Zhang
- Department of Internal Medicine, The Hospital of Traditional Chinese Medicine of Tai'an, Tai'an, Shandong 271000, P.R. China
| | - Helou Li
- Department of Laboratory, Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| | - Wei Zeng
- Department of Cardiology, Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| | - Mingqi Qiao
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| |
Collapse
|
16
|
Szymiczek A, Pastorino S, Larson D, Tanji M, Pellegrini L, Xue J, Li S, Giorgi C, Pinton P, Takinishi Y, Pass HI, Furuya H, Gaudino G, Napolitano A, Carbone M, Yang H. FTY720 inhibits mesothelioma growth in vitro and in a syngeneic mouse model. J Transl Med 2017; 15:58. [PMID: 28298211 PMCID: PMC5353897 DOI: 10.1186/s12967-017-1158-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/06/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Malignant mesothelioma (MM) is a very aggressive type of cancer, with a dismal prognosis and inherent resistance to chemotherapeutics. Development and evaluation of new therapeutic approaches is highly needed. Immunosuppressant FTY720, approved for multiple sclerosis treatment, has recently raised attention for its anti-tumor activity in a variety of cancers. However, its therapeutic potential in MM has not been evaluated yet. METHODS Cell viability and anchorage-independent growth were evaluated in a panel of MM cell lines and human mesothelial cells (HM) upon FTY720 treatment to assess in vitro anti-tumor efficacy. The mechanism of action of FTY720 in MM was assessed by measuring the activity of phosphatase protein 2A (PP2A)-a major target of FTY720. The binding of the endogenous inhibitor SET to PP2A in presence of FTY720 was evaluated by immunoblotting and immunoprecipitation. Signaling and activation of programmed cell death were evaluated by immunoblotting and flow cytometry. A syngeneic mouse model was used to evaluate anti-tumor efficacy and toxicity profile of FTY720 in vivo. RESULTS We show that FTY720 significantly suppressed MM cell viability and anchorage-independent growth without affecting normal HM cells. FTY720 inhibited the phosphatase activity of PP2A by displacement of SET protein, which appeared overexpressed in MM, as compared to HM cells. FTY720 promoted AKT dephosphorylation and Bcl-2 degradation, leading to induction of programmed cell death, as demonstrated by caspase-3 and PARP activation, as well as by cytochrome c and AIF intracellular translocation. Moreover, FTY720 administration in vivo effectively reduced tumor burden in mice without apparent toxicity. CONCLUSIONS Our preclinical data indicate that FTY720 is a potentially promising therapeutic agent for MM treatment.
Collapse
Affiliation(s)
- Agata Szymiczek
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Sandra Pastorino
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| | - David Larson
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Mika Tanji
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Laura Pellegrini
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Jiaming Xue
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Shuangjing Li
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Carlotta Giorgi
- Department of Morphology-Surgery-Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Morphology-Surgery-Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Yasutaka Takinishi
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, NY, 10065, USA
| | - Hideki Furuya
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Giovanni Gaudino
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Andrea Napolitano
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Michele Carbone
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| | - Haining Yang
- Thoracic Oncology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| |
Collapse
|