Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Drug Resist Updat ; 73: 101027, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38290407

ABSTRACT

AIMS: Pancreatic cancer (PC) is a highly metastatic malignant tumor of the digestive system. Drug resistance frequently occurs during cancer treatment process. This study aimed to explore the link between chemoresistance and tumor metastasis in PC and its possible molecular and cellular mechanisms. METHODS: A Metastasis and Chemoresistance Signature (MCS) scoring system was built and validated based on metastasis- and chemoresistance-related genes using gene expression data of PC, and the model was applied to single-cell RNA sequencing data. The influence of linker histone H1.2 (H1-2) on PC was explored through in vitro and in vivo experiments including proliferation, invasion, migration, drug sensitivity, rescue experiments and immunohistochemistry, emphasizing its regulation with c-MYC signaling pathway. RESULTS: A novel MCS scoring system accurately predicted PC patient survival and was linked to chemoresistance and epithelial-mesenchymal transition (EMT) in PC single-cell RNA sequencing data. H1-2 emerged as a significant prognostic factor, with its high expression indicating increased chemoresistance and EMT. This upregulation was mediated by c-MYC, which was also found to be highly expressed in PC tissues. CONCLUSION: The MCS scoring system offers insights into PC chemoresistance and metastasis potential. Targeting H1-2 could enhance therapeutic strategies and improve PC patient outcomes.


Subject(s)
Histones , Pancreatic Neoplasms , Humans , Histones/genetics , Histones/metabolism , Drug Resistance, Neoplasm/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/therapeutic use , Cell Line, Tumor , Signal Transduction , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic
2.
Clin Immunol ; 255: 109729, 2023 10.
Article in English | MEDLINE | ID: mdl-37562723

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease characterized by the immune system's failure to maintain self-tolerance, resulting in the autoimmune destruction of pancreatic beta cells. Although T1D has conventionally been viewed as a T-cell-dominant disease, recent research has emphasized the contribution of B cells in the onset of the disease. However, the mechanism underlying aberrant B cell responses remains unknown. B cell metabolism is a crucial prerequisite for B cell function and the development of adaptive immune responses. Here, we investigated the metabolic features of B cells, first in a cross-sectional cohort and subsequently in non-obese diabetic (NOD) mice, and revealed that there is an increased frequency of high-glucose-avidity (2-NBDGhigh) B cell population that may contribute to T1D progression. Further characterization of the metabolic, transcriptional and functional phenotype of B cells in NOD mice found that elevated glucose avidity is associated with a greater capacity for co-stimulation, proliferation and inflammatory cytokine production. Mechanistically, elevated Myc signaling orchestrated the glucose metabolism and the pro-inflammatory response of B cells in T1D. In vitro experiments demonstrated that pharmacological inhibition of glucose metabolism using metformin and 2-DG reduced pro-inflammatory cytokine production and B cell proliferation. Moreover, the combination of these inhibitors successfully delayed insulitis development, onset of diabetes, and improved high blood glucose levels in streptozotocin (STZ)-induced diabetic mice model. Taken together, our work has uncovered these high-glucose-avidity B cells as novel adjuvant diagnostic and therapeutic targets for T1D.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Humans , Mice , Animals , Mice, Inbred NOD , Cross-Sectional Studies , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/therapeutic use , Signal Transduction , Cytokines , Glucose
3.
Bone Marrow Transplant ; 58(9): 1000-1007, 2023 09.
Article in English | MEDLINE | ID: mdl-37198234

ABSTRACT

Dual expression of MYC and BCL2 proteins (double-expressor lymphoma [DEL]) as well as cell of origin (COO) are important prognostic factors in patients with diffuse large B-cell lymphoma (DLBCL) after conventional chemotherapy. We studied the prognostic impact of DEL and COO in patients with relapsed DLBCL treated with autologous stem cell transplant (ASCT). Three-hundred and three patients with stored tissue samples were identified. Classification was successful in 267 patients: 161 (60%) were DEL/non-double hit (DHL), 98 (37%) were non-DEL/non-DHL, and 8 (3%) were DEL/DHL. Compared to non-DEL/non-DHL, DEL/DHL had worse overall survival while DEL/non-DHL did not significantly differ in overall survival. On multivariable analysis, DEL/DHL, age >60 years, and >2 prior therapies, but not COO, were important prognostic factors for overall survival. When we explored the interaction of COO and BCL2 expression, patients with germinal center B-cell (GCB)/BCL2 (+) had inferior progression-free survival (PFS) compared to GCB/BCL2 (-) patients (HR, 4.97; P = 0.027). We conclude that the DEL/non-DHL and non-DEL/non-DHL subtypes of DLBCL have similar survival after ASCT. The negative impact of GCB/BCL2 (+) on PFS warrants future trials targeting BCL2 after ASCT. The inferior outcomes in DEL/DHL need to be verified in a larger number of patients.


Subject(s)
Clinical Relevance , Hematopoietic Stem Cell Transplantation , Lymphoma, Large B-Cell, Diffuse , Humans , Middle Aged , Autografts , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/analysis , Proto-Oncogene Proteins c-myc/therapeutic use
4.
Malays J Pathol ; 45(1): 1-10, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37119241

ABSTRACT

Lymphomas are prevalent worldwide and a common malignancy reported in Malaysia. Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of B-cell lymphomas accounting for 54% to 65% of all B-cell lymphomas and 39% to 57% of all malignant lymphomas. However, DLBCL comprises a heterogeneous group of diseases with different clinical presentations, biology and response to treatment. Recent advances in understanding the genetic landscape and molecular features of DLBCL have identified high-risk subsets with poor outcomes to chemo-immunotherapy that are actively being studied in various clinical trials. C-MYC is a proto-oncogene located in chromosome 8q24. 10 to 15 % of patients with newly diagnosed DLBCL have an underlying rearrangement of the MYC oncogene, resulting in dysregulated cellular survival and proliferation. Approximately half of these cases also carry a rearrangement of the anti-apoptotic proto-oncogene BCL2 and/or its transcription repressor BCL6. Over 20 case reports of DLBCL cases with notable features in Malaysia have found in the literature, in addition to a few extensive case series and included in this review. R-CHOP remains the mainstay of therapy and can help achieve control of long-term disease in nearly 90% of patients presenting with limited-stage and in up to 60% of those presenting with advanced stages. This review captures all 52 studies that reported DLBCL in Malaysia and summarises the essential aspects, including prevalence, subtype, prognostic markers clinical features in presentation and limited outcomes of cases when available.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Proto-Oncogene Proteins c-bcl-2 , Humans , Proto-Oncogene Proteins c-bcl-6/genetics , Malaysia , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/therapeutic use , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Prognosis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
5.
Drug Resist Updat ; 69: 100963, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37119690

ABSTRACT

MYC is a proto-oncogene that encodes a powerful regulator of transcription and cellular programs essential for normal development, as well as the growth and survival of various types of cancer cells. MYC rearrangement and amplification is a common cause of hematologic malignancies. In epithelial cancers such as colorectal cancer, genetic alterations in MYC are rare. Activation of Wnt, ERK/MAPK, and PI3K/mTOR pathways dramatically increases Myc levels through enhanced transcription, translation, and protein stability. Elevated Myc promotes stress adaptation, metabolic reprogramming, and immune evasion to drive cancer development and therapeutic resistance through broad changes in transcriptional and translational landscapes. Despite intense interest and effort, Myc remains a difficult drug target. Deregulation of Myc and its targets has profound effects that vary depending on the type of cancer and the context. Here, we summarize recent advances in the mechanistic understanding of Myc-driven oncogenesis centered around mRNA translation and proteostress. Promising strategies and agents under development to target Myc are also discussed with a focus on colorectal cancer.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins c-myc , Humans , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/therapeutic use , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology
6.
Endocrine ; 78(3): 446-457, 2022 12.
Article in English | MEDLINE | ID: mdl-36205915

ABSTRACT

PURPOSE: Sorafenib has been reported to reduce blood glucose levels in diabetic and non-diabetic patients in previous retrospective studies. However, the mechanism of which the hypoglycemic effects of sorafenib is not clearly explored. In this study, we investigated the effect of sorafenib on blood glucose levels in diabetic and normal mice and explored the possible mechanism. METHODS: We established a mouse model of type 2 diabetes by a high-fat diet combined with a low-dose of streptozotocin (STZ), to identify the hypoglycemic effect of sorafenib in different mice. Glucose tolerance, insulin tolerance and pyruvate tolerance tests were done after daily gavage with sorafenib to diabetic and control mice. To explore the molecular mechanism by which sorafenib regulates blood glucose levels, hepatic glucose metabolism signaling was studied by a series of in vivo and in vitro experiments. RESULTS: Sorafenib reduced blood glucose levels in both control and diabetic mice, particularly in the latter. The diabetic mice exhibited improved glucose and insulin tolerance after sorafenib treatment. Further studies showed that the expressions of gluconeogenesis-related enzymes, such as PCK1, G6PC and PCB, were significantly decreased upon sorafenib treatment. Mechanistically, sorafenib downregulates the expression of c-MYC downstream targets PCK1, G6PC and PCB through blocking the ERK/c-MYC signaling pathway, thereby playing its hypoglycemic effect by impairing hepatic glucose metabolism. CONCLUSION: Sorafenib reduces blood glucose levels through downregulating gluconeogenic genes, especially in diabetic mice, suggesting the patients with T2DM when treated with sorafenib need more emphasis in monitoring blood glucose to avoid unnecessary hypoglycemia.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Insulins , Mice , Animals , Blood Glucose/metabolism , Glucose/metabolism , Sorafenib/pharmacology , Sorafenib/therapeutic use , Diabetes Mellitus, Experimental/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/pharmacology , Proto-Oncogene Proteins c-myc/therapeutic use , Signal Transduction , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Liver/metabolism , Insulins/metabolism , Insulins/pharmacology , Insulins/therapeutic use , Insulin/metabolism
7.
Mol Metab ; 65: 101600, 2022 11.
Article in English | MEDLINE | ID: mdl-36113774

ABSTRACT

OBJECTIVE: Oral squamous cell carcinoma (OSCC) is characterized by high recurrence and metastasis and places a heavy burden on societies worldwide. Cancer cells thrive in a changing microenvironment by reprogramming lipidomic metabolic processes to provide nutrients and energy, activate oncogenic signaling pathways, and manage redox homeostasis to avoid lipotoxicity. The mechanism by which OSCC cells maintain lipid homeostasis during malignant progression is unclear. METHODS: The altered expression of fatty acid (FA) metabolism genes in OSCC, compared with that in normal tissues, and in OSCC patients with or without recurrence or metastasis were determined using public data from the TCGA and GEO databases. Immunohistochemistry was performed to examine the carboxylesterase 2 (CES2) protein level in our own cohort. CCK-8 and Transwell assays and an in vivo xenograft model were used to evaluate the biological functions of CES2. Mass spectrometry and RNA sequencing were performed to determine the lipidome and transcriptome alterations induced by CES2. Mitochondrial mass, mtDNA content, mitochondrial membrane potential, ROS levels, and oxygen consumption and apoptosis rates were evaluated to determine the effects of CES2 on mitochondrial function in OSCC. RESULTS: CES2 was downregulated in OSCC patients, especially those with recurrence or metastasis. CES2high OSCC patients showed better overall survival than CES2low OSCC patients. Restoring CES2 expression reduced OSCC cell viability and suppressed their migration and invasion in vitro, and it inhibited OSCC tumor growth in vivo. CES2 reprogrammed lipid metabolism in OSCC cells by hydrolyzing neutral lipid diacylglycerols (DGs) to release free fatty acids and reduce the membrane structure lipid phospholipids (PLs) synthesis. Free FAs were converted to acyl-carnitines (CARs) and transferred to mitochondria for oxidation, which induced reactive oxygen species (ROS) accumulation, mitochondrial damage, and apoptosis activation. Furthermore, the reduction in signaling lipids, e.g., DGs, PLs and substrates, suppressed PI3K/AKT/MYC signaling pathways. Restoring MYC rescued the diminished cell viability, suppressed migratory and invasive abilities, damaged mitochondria and reduced apoptosis rate induced by CES2. CONCLUSIONS: We demonstrated that CES2 downregulation plays an important role in OSCC by maintaining lipid homeostasis and reducing lipotoxicity during tumor progression and may provide a potential therapeutic target for OSCC.


Subject(s)
Carboxylesterase/metabolism , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Carboxylic Ester Hydrolases/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/pharmacology , DNA, Mitochondrial/therapeutic use , Diglycerides/metabolism , Fatty Acids, Nonesterified/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Homeostasis , Humans , Mitochondria/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/pharmacology , Proto-Oncogene Proteins c-myc/therapeutic use , Reactive Oxygen Species/metabolism , Signal Transduction , Sincalide/metabolism , Sincalide/pharmacology , Sincalide/therapeutic use , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
8.
Oncology (Williston Park) ; 36(8): 499-505, 2022 08 10.
Article in English | MEDLINE | ID: mdl-36001789

ABSTRACT

PURPOSE OF REVIEW: This article reviews the current data and future directions in the management of Burkitt lymphoma (BL) and high-grade B-cell lymphoma (HGBL). RECENT FINDINGS: BL is a rare, mature B-cell lymphoma molecularly defined by translocation of the proto-oncogene MYC. Multiple intensive combination chemoimmunotherapy regimens have demonstrated excellent efficacy in this disease, although treatment toxicity remains a challenge in many patients. Double-hit lymphoma (DHL) represents HGBL with translocations of the oncogene MYC along with either BCL2 or BCL6, or both. In 2016, the World Health Organization update of this classification was revised to a new entity defined by cytogenetics: HGBL with MYC and BCL2 and/or BCL6 rearrangements. Recent prospective data using dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab has demonstrated encouraging treatment efficacy in these patients. HGBL, not otherwise specified (NOS) is a heterogeneous, aggressive, mature B-cell lymphoma that does not meet criteria for BL, DHL, or diffuse large B-cell lymphoma NOS. Therapy for this entity is not well established. SUMMARY: The aggressive B-cell lymphomas BL, DHL, and HGBL, NOS are unique diseases with specific pathogenesis and biology. Insights into the molecular biology of these diseases have enabled new classifications and personalization of therapy.


Subject(s)
Burkitt Lymphoma , Lymphoma, Large B-Cell, Diffuse , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/therapeutic use , Translocation, Genetic
9.
J Zhejiang Univ Sci B ; 23(8): 666-681, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35953760

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is an aggressive type of non-Hodgkin's lymphoma. A total of 10%‒15% of DLBCL cases are associated with myelocytomatosis viral oncogene homolog(MYC) and/or B-cell lymphoma-2 (BCL2) translocation or amplification. BCL2 inhibitors have potent anti-tumor effects in DLBCL; however, resistance can be acquired through up-regulation of alternative anti-apoptotic proteins. The histone deacetylase (HDAC) inhibitor chidamide can induce BIM expression, leading to apoptosis of lymphoma cells with good efficacy in refractory recurrent DLBCL. In this study, the synergistic mechanism of chidamide and venetoclax in DLBCL was determined through in vitro and in vivo models. We found that combination therapy significantly reduced the protein levels of MYC, TP53, and BCL2 in activated apoptotic-related pathways in DLBCL cells by increasing BIM levels and inducing cell apoptosis. Moreover, combination therapy regulated expression of multiple transcriptomes in DLBCL cells, involving apoptosis, cell cycle, phosphorylation, and other biological processes, and significantly inhibited tumor growth in DLBCL-bearing xenograft mice. Taken together, these findings verify the in vivo therapeutic potential of chidamide and venetoclax combination therapy in DLBCL, warranting pre-clinical trials for patients with DLBCL.


Subject(s)
Biological Phenomena , Lymphoma, Large B-Cell, Diffuse , Aminopyridines , Animals , Benzamides , Bridged Bicyclo Compounds, Heterocyclic , Down-Regulation , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Neoplasm Recurrence, Local , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/therapeutic use , Sulfonamides , Tumor Suppressor Protein p53/metabolism
10.
Life Sci Alliance ; 5(9)2022 09.
Article in English | MEDLINE | ID: mdl-35500936

ABSTRACT

Mutations in the CFTR anion channel cause cystic fibrosis (CF) and have also been related to higher cancer incidence. Previously we proposed that this is linked to an emerging role of functional CFTR in protecting against epithelial-mesenchymal transition (EMT). However, the pathways bridging dysfunctional CFTR to EMT remain elusive. Here, we applied systems biology to address this question. Our data show that YAP1 is aberrantly active in the presence of mutant CFTR, interacting with F508del, but not with wt-CFTR, and that YAP1 knockdown rescues F508del-CFTR processing and function. Subsequent analysis of YAP1 interactors and roles in cells expressing either wt- or F508del-CFTR reveal that YAP1 is an important mediator of the fibrotic/EMT processes in CF. Alongside, five main pathways emerge here as key in linking mutant CFTR to EMT, namely, (1) the Hippo pathway; (2) the Wnt pathway; (3) the TGFß pathway; (4) the p53 pathway; and (5) MYC signaling. Several potential hub proteins which mediate the crosstalk among these pathways were also identified, appearing as potential therapeutic targets for both CF and cancer.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial-Mesenchymal Transition/genetics , Humans , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/therapeutic use , Signal Transduction/genetics , YAP-Signaling Proteins
11.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799592

ABSTRACT

B-Cell Lymphoma 2 (BCL-2), c-MYC and related proteins are arguably amongst the most widely studied in all of biology. Every year there are thousands of papers reporting on different aspects of their biochemistry, cellular and physiological mechanisms and functions. This plethora of literature can be attributed to both proteins playing essential roles in the normal functioning of a cell, and by extension a whole organism, but also due to their central role in disease, most notably, cancer. Many cancers arise due to genetic lesions resulting in deregulation of both proteins, and indeed the development and survival of tumours is often dependent on co-operativity between these protein families. In this review we will discuss the individual roles of both proteins in cancer, describe cancers where co-operativity between them has been well-characterised and finally, some strategies to target these proteins therapeutically.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/genetics , Aniline Compounds/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Biphenyl Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Clinical Trials as Topic , Gene Expression Regulation, Neoplastic , Humans , Morpholinos/therapeutic use , Neoplasms/metabolism , Neoplasms/pathology , Nitrophenols/therapeutic use , Peptide Fragments/therapeutic use , Piperazines/therapeutic use , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/therapeutic use , Rituximab/therapeutic use , Signal Transduction , Sulfonamides/therapeutic use
12.
Arch Med Res ; 51(7): 636-644, 2020 10.
Article in English | MEDLINE | ID: mdl-32553459

ABSTRACT

BACKGROUNDS: Although ATO is widely used to treat acute promelocytic leukemia (APL), the appropriate effects of the drug as a single agent are achieved in high doses which are not clinically achievable without the risk of side effects; highlighting the necessity of its application in a combined-modality. Herein, we aimed to investigate whether c-Myc inhibition could reinforce the anti-leukemic effect of ATO, while reducing its concentration in APL cells. METHODS: NB4 cells were treated with the relevant concentrations of 10058-F4 (c-Myc inhibitor) and ATO, and then the survival of the cells was evaluated using trypan blue, MTT and BrdU assays. Moreover, the mechanism of action of the agents were evaluated using Flow cytometry, qRT-PCR and western blot analysis. RESULTS: We found that the inhibition of c-Myc using 10058-F4 could enhance the anti-leukemic effect of ATO in APL cells through reducing the phosphorylation of IκB, decreasing the expression of the anti-apoptotic genes and in turn, inducing a caspase-3-dependent apoptotic cell death. Moreover, the combination of 10058-F4 and ATO abrogated the activation of the PI3K pathway, while neither agent had significant suppressive impact on this pathway; suggesting for the first time that probably the companionship of c-Myc inhibitor may be an appealing strategy for shifting the resistance condition toward a chemo-sensitive phenotype, without the necessity to elevate the effective dose of ATO. CONCLUSION: Given the efficacy of 10058-F4 in adjuvanting approaches, we suggest this small molecule inhibitor as an impressing agent to be used alongside ATO in the treatment of APL.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenic Trioxide/therapeutic use , Genes, myc/genetics , Leukemia, Promyelocytic, Acute/drug therapy , NF-kappa B/metabolism , Proto-Oncogene Proteins c-myc/therapeutic use , Thiazoles/therapeutic use , Antineoplastic Agents/pharmacology , Arsenic Trioxide/pharmacology , Cell Line, Tumor , Humans , Proto-Oncogene Proteins c-myc/pharmacology , Thiazoles/pharmacology
13.
Sci Transl Med ; 11(484)2019 03 20.
Article in English | MEDLINE | ID: mdl-30894502

ABSTRACT

Inhibiting MYC has long been considered unfeasible, although its key role in human cancers makes it a desirable target for therapeutic intervention. One reason for its perceived undruggability was the fear of catastrophic side effects in normal tissues. However, we previously designed a dominant-negative form of MYC called Omomyc and used its conditional transgenic expression to inhibit MYC function both in vitro and in vivo. MYC inhibition by Omomyc exerted a potent therapeutic impact in various mouse models of cancer, causing only mild, well-tolerated, and reversible side effects. Nevertheless, Omomyc has been so far considered only a proof of principle. In contrast with that preconceived notion, here, we show that the purified Omomyc mini-protein itself spontaneously penetrates into cancer cells and effectively interferes with MYC transcriptional activity therein. Efficacy of the Omomyc mini-protein in various experimental models of non-small cell lung cancer harboring different oncogenic mutation profiles establishes its therapeutic potential after both direct tissue delivery and systemic administration, providing evidence that the Omomyc mini-protein is an effective MYC inhibitor worthy of clinical development.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/therapeutic use , DNA/metabolism , Disease Models, Animal , E-Box Elements/genetics , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Peptide Fragments/therapeutic use , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-myc/administration & dosage , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/pharmacokinetics , Proto-Oncogene Proteins c-myc/pharmacology , Proto-Oncogene Proteins c-myc/therapeutic use
14.
Oncogene ; 38(1): 140-150, 2019 01.
Article in English | MEDLINE | ID: mdl-30076412

ABSTRACT

Overexpression of MYC oncogene is highly prevalent in many malignancies such as aggressive triple-negative breast cancers (TNBCs) and it is associated with very poor outcome. Despite decades of research, attempts to effectively inhibit MYC, particularly with small molecules, still remain challenging due to the featureless nature of its protein structure. Herein, we describe the engineering of the dominant-negative MYC peptide (OmoMYC) linked to a functional penetrating 'Phylomer' peptide (FPPa) as a therapeutic strategy to inhibit MYC in TNBC. We found FPPa-OmoMYC to be a potent inducer of apoptosis (with IC50 from 1-2 µM) in TNBC cells with negligible effects in non-tumorigenic cells. Transcriptome analysis of FPPa-OmoMYC-treated cells indicated that the fusion protein inhibited MYC-dependent networks, inducing dynamic changes in transcriptional, metabolic, and apoptotic processes. We demonstrated the efficacy of FPPa-OmoMYC in inhibiting breast cancer growth when injected orthotopically in TNBC allografts. Lastly, we identified strong pharmacological synergisms between FPPa-OmoMYC and chemotherapeutic agents. This study highlights a novel therapeutic approach to target highly aggressive and chemoresistant MYC-activated cancers.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Molecular Targeted Therapy/methods , Neoplasm Proteins/antagonists & inhibitors , Peptide Fragments/therapeutic use , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Amino Acid Sequence , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Female , Genes, myc , Humans , Inhibitory Concentration 50 , Leucine Zippers/genetics , Mice , Models, Molecular , Mutation , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/pharmacokinetics , Peptide Library , Protein Conformation , Protein Engineering , Proto-Oncogene Proteins c-myc/administration & dosage , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/pharmacokinetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics
15.
Biomaterials ; 65: 43-55, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26142775

ABSTRACT

Tumor cell nucleus-targeted delivery of antitumor agents is of great interest in cancer therapy, since the nucleus is one of the most frequent targets of drug action. Here we report a smart polymeric conjugate platform, which utilizes stimulus-responsive strategies to achieve multistage nuclear drug delivery upon systemic administration. The conjugates composed of a backbone based on N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer and detachable nucleus transport sub-units that sensitive to lysosomal enzyme. The sub-units possess a biforked structure with one end conjugated with the model drug, H1 peptide, and the other end conjugated with a novel pH-responsive targeting peptide (R8NLS) that combining the strength of cell penetrating peptide and nuclear localization sequence. The conjugates exhibited prolonged circulation time and excellent tumor homing ability. And the activation of R8NLS in acidic tumor microenvironment facilitated tissue penetration and cellular internalization. Once internalized into the cell, the sub-units were unleashed for nuclear transport through nuclear pore complex. The unique features resulted in 50-fold increase of nuclear drug accumulation relative to the original polymer-drug conjugates in vitro, and excellent in vivo nuclear drug delivery efficiency. Our report provides a strategy in systemic nuclear drug delivery by combining the microenvironment-responsive structure and detachable sub-units.


Subject(s)
Acrylamides/chemistry , Antineoplastic Agents/administration & dosage , Cell Nucleus/metabolism , Cell-Penetrating Peptides/chemistry , Delayed-Action Preparations/chemistry , Peptides/administration & dosage , Proto-Oncogene Proteins c-myc/administration & dosage , Acrylamides/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell-Penetrating Peptides/metabolism , Delayed-Action Preparations/metabolism , Drug Delivery Systems , HeLa Cells , Humans , Hydrogen-Ion Concentration , Male , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasms/drug therapy , Neoplasms/pathology , Nuclear Localization Signals , Peptides/chemistry , Peptides/pharmacokinetics , Peptides/therapeutic use , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/pharmacokinetics , Proto-Oncogene Proteins c-myc/therapeutic use
16.
Genes Dev ; 27(5): 504-13, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23475959

ABSTRACT

The principal reason for failure of targeted cancer therapies is the emergence of resistant clones that regenerate the tumor. Therapeutic efficacy therefore depends on not only how effectively a drug inhibits its target, but also the innate or adaptive functional redundancy of that target and its attendant pathway. In this regard, the Myc transcription factors are intriguing therapeutic targets because they serve the unique and irreplaceable role of coordinating expression of the many diverse genes that, together, are required for somatic cell proliferation. Furthermore, Myc expression is deregulated in most-perhaps all-cancers, underscoring its irreplaceable role in proliferation. We previously showed in a preclinical mouse model of non-small-cell lung cancer that systemic Myc inhibition using the dominant-negative Myc mutant Omomyc exerts a dramatic therapeutic impact, triggering rapid regression of tumors with only mild and fully reversible side effects. Using protracted episodic expression of Omomyc, we now demonstrate that metronomic Myc inhibition not only contains Ras-driven lung tumors indefinitely, but also leads to their progressive eradication. Hence, Myc does indeed serve a unique and nondegenerate role in lung tumor maintenance that cannot be complemented by any adaptive mechanism, even in the most aggressive p53-deficient tumors. These data endorse Myc as a compelling cancer drug target.


Subject(s)
Lung Neoplasms/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/pharmacology , Proto-Oncogene Proteins c-myc/therapeutic use , Survival Analysis , Tumor Suppressor Protein p53/metabolism
17.
PLoS One ; 6(7): e22284, 2011.
Article in English | MEDLINE | ID: mdl-21811581

ABSTRACT

Recent evidence points to Myc--a multifaceted bHLHZip transcription factor deregulated in the majority of human cancers--as a priority target for therapy. How to target Myc is less clear, given its involvement in a variety of key functions in healthy cells. Here we report on the action mechanism of the Myc interfering molecule termed Omomyc, which demonstrated astounding therapeutic efficacy in transgenic mouse cancer models in vivo. Omomyc action is different from the one that can be obtained by gene knockout or RNA interference, approaches designed to block all functions of a gene product. This molecule--instead--appears to cause an edge-specific perturbation that destroys some protein interactions of the Myc node and keeps others intact, with the result of reshaping the Myc transcriptome. Omomyc selectively targets Myc protein interactions: it binds c- and N-Myc, Max and Miz-1, but does not bind Mad or select HLH proteins. Specifically, it prevents Myc binding to promoter E-boxes and transactivation of target genes while retaining Miz-1 dependent binding to promoters and transrepression. This is accompanied by broad epigenetic changes such as decreased acetylation and increased methylation at H3 lysine 9. In the presence of Omomyc, the Myc interactome is channeled to repression and its activity appears to switch from a pro-oncogenic to a tumor suppressive one. Given the extraordinary therapeutic impact of Omomyc in animal models, these data suggest that successfully targeting Myc for cancer therapy might require a similar twofold action, in order to prevent Myc/Max binding to E-boxes and, at the same time, keep repressing genes that would be repressed by Myc.


Subject(s)
Molecular Targeted Therapy , Neoplasms/drug therapy , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Proteins/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , Down-Regulation/genetics , Epigenesis, Genetic , Fibroblasts/metabolism , Humans , Intracellular Space/metabolism , Mice , Neoplasms/pathology , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/pharmacology , Proto-Oncogene Proteins c-myc/therapeutic use , Rats , Repressor Proteins/metabolism , Serum , Transcription, Genetic , Transcriptional Activation/genetics , Up-Regulation/genetics
18.
Int J Biochem Cell Biol ; 36(5): 771-5, 2004 May.
Article in English | MEDLINE | ID: mdl-15061133

ABSTRACT

N-myc has emerged as a member of a transcriptional regulatory network which impinges directly on the machinery of cell growth and proliferation. Critical during neural crest embryogenesis, N-myc is rapidly down-regulated as tissues become terminally differentiated and growth-arrested. The involvement of N-myc in these fundamental cellular processes necessitates an intricate strategy for its regulation, which is still being elucidated. Deregulated N-myc over-expression has clear transforming ability in vitro and in vivo. The transcriptional target genes responsible for this activity are beginning to be unravelled.


Subject(s)
Proto-Oncogene Proteins c-myc/physiology , Transcription Factors/physiology , Animals , Gene Expression Regulation , Humans , Mice , Neural Crest/embryology , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/therapeutic use , Transcription Factors/genetics , Transcription Factors/therapeutic use
19.
Cardiovasc Radiat Med ; 3(1): 26-30, 2002.
Article in English | MEDLINE | ID: mdl-12479913

ABSTRACT

BACKGROUND: Neointimal hyperplasia following percutaneous transluminal coronary angioplasty (PTCA) is one of the major components of the process of restenosis. We evaluated the long-term impact of local delivery of c-myc neutrally charged antisense oligonucleotides (Resten-NG) upon neointimal formation following PTCA in a rabbit model. METHODS: PTCA was performed in the iliac arteries of 10 New Zealand white rabbits at 8 atm for 30 s, three times. An infusion of 500 micro g Resten-NG (n=6) or saline (n=4) was delivered to the site at 2 atm via the outer balloon pores of the transport catheter over 2 min. The diet was supplemented with 0.25% cholesterol for 10 days before and 6 months following PTCA. RESULTS: After 6 months, animals were sacrificed and vessels were fixed in formalin, processed and stained with hematoxylin, eosin, and movat. Histological analysis revealed complete vascular healing in both groups of animals. Planimetry showed that intimal areas were 1.71+/-0.25 and 0.65+/-0.36 mm2 in the control and antisense delivery groups, respectively (P<.05). CONCLUSION: We conclude that local delivery of Resten-NG significantly inhibited neointimal thickening following PTCA in a rabbit for up to 6 months.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Oligodeoxyribonucleotides, Antisense/therapeutic use , Postoperative Complications/drug therapy , Postoperative Complications/etiology , Proto-Oncogene Proteins c-myc/therapeutic use , Tunica Intima/injuries , Tunica Intima/pathology , Vascular Diseases/drug therapy , Vascular Diseases/etiology , Wound Healing/physiology , Animals , Disease Models, Animal , Drug Delivery Systems , Extracellular Matrix/metabolism , Fibrin/metabolism , Follow-Up Studies , Iliac Artery/injuries , Iliac Artery/metabolism , Iliac Artery/surgery , Myocytes, Smooth Muscle/metabolism , Necrosis , Postoperative Complications/metabolism , Rabbits , Severity of Illness Index , Time , Treatment Outcome , Tunica Intima/metabolism , Vascular Diseases/metabolism
20.
Exp Eye Res ; 66(5): 581-9, 1998 May.
Article in English | MEDLINE | ID: mdl-9628805

ABSTRACT

The purpose of this work was to investigate if MYC-dependent intracellular mitogenic pathway is active in cultures of human retinal pigment epithelial (hRPE) cells and whether myc antisense phosphorotioate oligonucleotides (c-myc-AS-ODN) are useful tools for inhibiting the proliferation of hRPE cells. Cultures of hRPE cells were established from adult human corneal donors. These cells were positively stained for cytokeratins and vimentin. Myc mRNA expression was determined by Northern blot analysis and it was determined by means of immunofluorescence if MYC was expressed. C-myc-AS-ODN effect on cell proliferation was estimated by evaluating the incorporation of 5-bromo-2'-deoxy-uridine into cellular DNA. Cell number was estimated by using a tetrazolium bromide based colorimetric method. Human RPE cells in culture expressed MYC and myc mRNA as well as prothymosin alpha mRNA--a gene whose transcription is under MYC control--indicating that MYC-dependent intracellular mitogenic pathway is active in these cells. In accordance with this, we found that blocking the expression of myc by the addition of c-myc-AS-ODN to the culture medium inhibited hRPE cell proliferation. The effect of the c-myc-AS-ODN was found to be sequence specific (the use of a control oligonucleotide with the same sequence but in an opposite direction had no effect) and dose-dependent (4 microM was the lowest effective dose tested). By using RT-PCR we found that the c-myc-AS-ODN inhibition of cell proliferation was related to a diminution in c-myc mRNA expression, and by immunofluorescence we detected a diminution in c-MYC protein staining in RPE cells after 48 hr of treatment with c-myc-AS-ODN. Furthermore, growth inhibition remained for at least 5 days after addition of a single dose of the c-myc-AS-ODN to the culture. We conclude that hRPE cell proliferation is under MYC control. Blocking the expression of myc by c-myc-AS-ODN inhibited hRPE cell proliferation. These findings establish a rationale for investigating the potential use of a c-myc-AS-ODN as a novel therapeutical tool in the treatment of Proliferative Vitreoretinopathy.


Subject(s)
Oligonucleotides, Antisense/metabolism , Pigment Epithelium of Eye/cytology , Proto-Oncogene Proteins c-myc/metabolism , Adult , Blotting, Northern , Cell Division/genetics , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression , Humans , Keratins/metabolism , Oligonucleotides, Antisense/therapeutic use , Polymerase Chain Reaction/methods , Proto-Oncogene Proteins c-myc/therapeutic use , RNA, Messenger/metabolism , Vimentin/metabolism , Vitreoretinopathy, Proliferative/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...