1
|
Vesović M, Jelić R, Nikolić M, Nedeljković N, Živanović A, Bukonjić A, Mrkalić E, Radić G, Ratković Z, Kljun J, Tomović D. Investigation of the interaction between S-isoalkyl derivatives of the thiosalicylic acid and human serum albumin. J Biomol Struct Dyn 2025; 43:4081-4094. [PMID: 38192057 DOI: 10.1080/07391102.2024.2301745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/30/2023] [Indexed: 01/10/2024]
Abstract
S-isoalkyl derivatives of thiosalicylic acid (isopropyl-(L1), isobutyl-(L2) and isoamyl-(L3)) were selected in order to investigate the binding interaction with the human serum albumin (HSA) using different spectroscopic methods and molecular docking simulation. Association constants and number of binding sites were used to analyze the quenching mechanism. The experimental results showed that the fluorescence quenching of HSA by L1, L2 and L3 occurs because of static quenching and that binding processes were spontaneous, with the leading forces in bonding by hydrogen bonding, hydrophobic interactions, and electrostatic interactions. Fluorescence spectroscopy, UV-Vis spectroscopy and synchronous fluorescence spectroscopy showed that ligands (L1, L2 and L3) can bind to HSA and that the binding of ligands induced some microenvironmental and conformational changes in HSA. The calculated distance between the donor and the acceptor according to fiFörster's theory confirms the energy transfer efficiency between the acceptor and HSA. Results of site marker competitive experiments showed that the tested compounds bind to HSA in domain IIA (Site I). Molecular dynamics and docking calculations demonstrated that L3 binds to the Sudlow site I of HSA with lower values of binding energies compared to L1 and L2, indicating the formation of the most stable ligand-HSA complex. Understanding the binding mechanisms of S-isoalkyl derivatives of the thiosalicylic acid to HSA may provide valuable data for the future studies of their biological activity and application as potential antitumor drugs.
Collapse
Affiliation(s)
- Marina Vesović
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, Kragujevac, Serbia
| | - Ratomir Jelić
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, Kragujevac, Serbia
| | - Miloš Nikolić
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, Kragujevac, Serbia
| | - Nikola Nedeljković
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, Kragujevac, Serbia
| | - Ana Živanović
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, Kragujevac, Serbia
| | - Andriana Bukonjić
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, Kragujevac, Serbia
| | - Emina Mrkalić
- University of Kragujevac, Institute for Information Technologies, Department of Science, Jovana Cvijića bb, Kragujevac, Serbia
| | - Gordana Radić
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, Kragujevac, Serbia
| | - Zoran Ratković
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, Kragujevac, Serbia
| | - Jakob Kljun
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Večna pot 113, Ljubljana, Slovenia
| | - Dušan Tomović
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, Kragujevac, Serbia
| |
Collapse
|
2
|
Chen Y, Yin Z, Westover KD, Zhou Z, Shu L. Advances and Challenges in RAS Signaling Targeted Therapy in Leukemia. Mol Cancer Ther 2025; 24:33-46. [PMID: 39404173 DOI: 10.1158/1535-7163.mct-24-0504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/04/2024] [Accepted: 10/08/2024] [Indexed: 01/03/2025]
Abstract
RAS mutations are prevalent in leukemia, including mutations at G12, G13, T58, Q61, K117, and A146. These mutations are often crucial for tumor initiation, maintenance, and recurrence. Although much is known about RAS function in the last 40 years, a substantial knowledge gap remains in understanding the mutation-specific biological activities of RAS in cancer and the approaches needed to target specific RAS mutants effectively. The recent approval of KRASG12C inhibitors, adagrasib and sotorasib, has validated KRAS as a direct therapeutic target and demonstrated the feasibility of selectively targeting specific RAS mutants. Nevertheless, KRASG12C remains the only RAS mutant successfully targeted with FDA-approved inhibitors for cancer treatment in patients, limiting its applicability for other oncogenic RAS mutants, such as G12D, in leukemia. Despite these challenges, new approaches have generated optimism about targeting specific RAS mutations in an allele-dependent manner for cancer therapy, supported by compelling biochemical and structural evidence, which inspires further exploration of RAS allele-specific vulnerabilities. This review will discuss the recent advances and challenges in the development of therapies targeting RAS signaling, highlight emerging therapeutic strategies, and emphasize the importance of allele-specific approaches for leukemia treatment.
Collapse
Affiliation(s)
- Yu Chen
- Department of Immunology, Guizhou Province Key Laboratory for Regenerative Medicine, Clinical Research Center, School of Basic Medicine, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Zhenghao Yin
- Department of Immunology, Guizhou Province Key Laboratory for Regenerative Medicine, Clinical Research Center, School of Basic Medicine, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Kenneth D Westover
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Zhiwei Zhou
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Liping Shu
- Department of Immunology, Guizhou Province Key Laboratory for Regenerative Medicine, Clinical Research Center, School of Basic Medicine, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
- Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China
| |
Collapse
|
3
|
Kim DE, Oh HJ, Kim HJ, Kim YB, Kim ST, Yim H. Synergistic two-step inhibition approach using a combination of trametinib and onvansertib in KRAS and TP53-mutated colorectal adenocarcinoma. Biomed Pharmacother 2025; 182:117796. [PMID: 39731938 DOI: 10.1016/j.biopha.2024.117796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024] Open
Abstract
Colorectal malignancies associated with KRAS and TP53 mutations led us to investigate the effects of combination therapy targeting KRAS, MEK1, or PLK1 in colorectal cancer. MEK1 is downstream of RAS in the MAPK pathway, whereas PLK1 is a mitotic kinase of the cell cycle activated by MAPK and regulated by p53. Bioinformatics analysis revealed that patients with colorectal cancer had a high expression of MAP2K1 and PLK1. Furthermore, PLK1 and MEK1 activity in human colorectal adenocarcinoma (COAD) tissues was found to be highly upregulated compared to healthy tissues. To determine the sensitivity of KRAS or/and TP53-mutated cancer to KRAS, MEK1, or PLK1-targeted therapy, the inhibitors salirasib, trametinib, volasertib, and onvansertib were used in COAD cells with different KRAS and TP53 status. The results showed that combinations with trametinib and PLK1 inhibitors were more potent than combinations with salirasib. A combination of MEK1 and PLK1 inhibitors exhibited significant therapeutic effects on KRAS or/and TP53-mutated COAD cells. Notably, the combination of trametinib and onvansertib effectively suppressed tumor growth in a xenograft mouse model of KRAS and TP53-mutated COAD. This treatment induced G1 and G2/M arrest, respectively, and showed the strongest synergistic effect in KRAS and TP53-mutated SW48 cells expressing mutant KRASG13D and transduced with TP53 shRNA, ultimately leading to apoptotic cell death. These effects are attributed to two-step inhibition mechanism that blocks the MAPK signaling pathway and disrupts mitosis in KRAS and TP53-mutated COAD cells.
Collapse
Affiliation(s)
- Da-Eun Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hyun-Ji Oh
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hyun-Jin Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Yeo-Bin Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Seung-Tae Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea.
| |
Collapse
|
4
|
Sadeghi Shaker M, Rokni M, Kavosi H, Enayati S, Madreseh E, Mahmoudi M, Farhadi E, Vodjgani M. Salirasib Inhibits the Expression of Genes Involved in Fibrosis in Fibroblasts of Systemic Sclerosis Patients. Immun Inflamm Dis 2024; 12:e70063. [PMID: 39601641 PMCID: PMC11600624 DOI: 10.1002/iid3.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/01/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Fibrosis is a principal sign of systemic sclerosis (SSc) which can affect several organs including the lung, heart, and dermis. Dermal fibroblasts of SSc patients are characterized by persistent and activated Ras and ERK1/2 signaling which stimulates extreme collagen and extracellular matrix synthesis. Salirasib is a Ras inhibitor that competitively prevents the adherence of GTP-bound Ras to the plasma membrane, that inhibits Ras signaling. This study intended to clarify whether salirasib can influence fibrotic mediators in SSc fibroblasts. MATERIALS AND METHODS Dermal fibroblasts from 10 SSc patients were treated with salirasib in the presence of TGF-β1, and mRNA levels of H-Ras and genes related to fibrosis, such as COL1A1, COL1A2, CTGF, TGF-β1, fibronectin, ACTA2, and MMP1 was measured by real-time PCR. The α-SMA protein expression was analyzed by immunofluorescence staining. RESULTS In dermal fibroblasts of SSc patients, salirasib treatment, markedly downregulated the H-Ras gene expression. In addition, the protein expression of α-SMA and gene expression of ACTA2 were inhibited upon salirasib treatment. Salirasib also significantly reduced the expression of COL1A1, and COL1A2 genes and augmented the gene expression of MMP1. The mRNA levels of other genes related to fibrosis such as FN1, CTGF, and TGF-β1 were significantly decreased upon salirasib treatment. CONCLUSION Considering salirasib significantly reduced the expression of genes related to the fibrosis process and α-SMA gene and protein expression, and given significant upregulation of MMP1 by salirasib, it can be considered as a new curative strategy for fibrotic diseases like SSc.
Collapse
Affiliation(s)
- Mina Sadeghi Shaker
- Department of Immunology, School of MedicineTehran University of Medical SciencesTehranIran
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
| | - Mohsen Rokni
- Department of Immunology, School of MedicineTehran University of Medical SciencesTehranIran
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Department of ImmunologyUniversity of social Welfare and Rehabilitation SciencesTehranIran
| | - Hoda Kavosi
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Research Center for Chronic Inflammatory DiseasesTehran University of Medical SciencesTehranIran
| | - Samaneh Enayati
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
| | - Elham Madreseh
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Department of Epidemiology and Biostatistics, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Mahdi Mahmoudi
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Research Center for Chronic Inflammatory DiseasesTehran University of Medical SciencesTehranIran
| | - Elham Farhadi
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Research Center for Chronic Inflammatory DiseasesTehran University of Medical SciencesTehranIran
| | - Mohammad Vodjgani
- Department of Immunology, School of MedicineTehran University of Medical SciencesTehranIran
| |
Collapse
|
5
|
Gong AY, Qiao YJ, Chen M, Alam Z, Malhotra DK, Dworkin L, Ju W, Gunning WT. Glomerular injury induced by vinyl carbamate in A/J inbred mice: a novel model of membranoproliferative glomerulonephritis. Front Pharmacol 2024; 15:1462936. [PMID: 39309006 PMCID: PMC11412833 DOI: 10.3389/fphar.2024.1462936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
Ethyl carbamate (EC) is a process contaminant found in fermented foods and alcoholic beverages. Metabolic conversion of ethyl carbamate generates vinyl carbamate (VC), a carcinogenic metabolite. EC, as a Group 2A probable human carcinogen, and the more potent VC, are known to cause tumors in rodents. However, their effects on the kidney are unknown and were explored here. Female A/J inbred mice received an intraperitoneal injection of vehicle or VC. Beginning 5 weeks after VC injection, mice showed signs of moribund state. Mouse necropsies revealed renal glomerular injury that histopathologically recapitulated human membranoproliferative glomerulonephritis (MPGN), as evidenced by light microscopy, immunostaining for immunoglobulins and complements, and electron microscopy. To determine the molecular pathomechanisms, a post-hoc analysis was performed on a publicly available RNA-Seq transcriptome of kidneys from control rats and rats treated with fermented wine containing high concentrations of EC. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses of the differentially expressed genes revealed that the complement and coagulation cascades were a top predicted biological process involved. Furthermore, pathway-based data integration and visualization revealed that key regulators of complement activation were altered by high EC treatment. Among these, complement factors (CF) D and H, critical positive and negative regulators of the alternative pathway, respectively, were most affected, with CFD induced by 3.49-fold and CFH repressed by 5.9-fold, underscoring a hyperactive alternative pathway. Consistently, exposure of primary glomerular endothelial cells to EC or VC resulted in induction of CFD and repression of CFH, accompanied by increased fixation of C3 and C5b9. This effect seems to be mediated by Ras, one of the top genes that interact with both EC and VC, as identified by analyzing the chemical-gene/protein interactions database. Indeed, EC or VC-elicited complement activation was associated with activation of Ras signaling, but was abolished by the Ras inhibitor farnesyl thiosalicylic acid. Collectively, our findings suggest that VC, a metabolite of EC, induces glomerular injury in mice akin to human MPGN, possibly via perturbing the expression of complement regulators, resulting in an effect that favors activation of the alternative complement pathway.
Collapse
Affiliation(s)
- Athena Y. Gong
- College of Literature, Science and the Arts, University of Michigan, Ann Arbor, MI, United States
- Michigan O’Brien Kidney National Resource Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, United States
| | - Ying Jin Qiao
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, United States
| | - Mengxuan Chen
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, United States
| | - Zubia Alam
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, United States
| | - Deepak K. Malhotra
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, United States
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, United States
| | - Wenjun Ju
- Michigan O’Brien Kidney National Resource Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - William T. Gunning
- Department of Pathology, Medical College of Ohio at University of Toledo, Toledo, OH, United States
| |
Collapse
|
6
|
Lee EJ, Charles JF, Sinha I, Neppl RL. Loss of HNRNPU in Skeletal Muscle Increases Intramuscular Infiltration of Ly6C Positive Cells, leading to Muscle Atrophy through Activation of NF-κB Signaling. Adv Biol (Weinh) 2024; 8:e2400152. [PMID: 38797891 DOI: 10.1002/adbi.202400152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/10/2024] [Indexed: 05/29/2024]
Abstract
Heterogeneous nuclear ribonucleoprotein U (hnRNPU) is known to play multiple biological roles by regulating transcriptional expression, RNA splicing, RNA stability, and chromatin structure in a tissue-dependent manner. The role of hnRNPU in skeletal muscle development and maintenance has not been previously evaluated. In this study, skeletal muscle specific hnRNPU knock out mice is utilized and evaluated skeletal muscle mass and immune cell infiltration through development. By 4 weeks, muscle-specific hnRNPU knockout mice revealed Ly6C+ monocyte infiltration into skeletal muscle, which preceded muscle atrophy. Canonical NF-kB signaling is activated in a myofiber-autonomous manner with hnRNPU repression. Inducible hnRNPU skeletal muscle knockout mice further demonstrated that deletion of hnRNPU in adulthood is sufficient to cause muscle atrophy, suggesting that hnRNPU's role in muscle maintenance is not during development alone. Treatment with salirasib, to inhibit proliferation of immune cells, prevents muscle atrophy in muscle-specific hnRNPU knock out mice, indicating that immune cell infiltration plays causal role in muscle atrophy of hnRNPU knock out mice. Overall, the findings suggest that loss of hnRNPU triggers muscle inflammation and activates NF-κB signaling in a cell-autonomous manner, culminating in muscle atrophy.
Collapse
Affiliation(s)
- Eun-Joo Lee
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Julia F Charles
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Indranil Sinha
- Division of Plastic and reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Ronald L Neppl
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
7
|
Lim MW, Quan Tang Y, Aroua MK, Gew LT. Glycerol Extraction of Bioactive Compounds from Thanaka ( Hesperethusa crenulata) Bark through LCMS Profiling and Their Antioxidant Properties. ACS OMEGA 2024; 9:14388-14405. [PMID: 38559928 PMCID: PMC10976408 DOI: 10.1021/acsomega.4c00041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024]
Abstract
Organic solvents are hazardous to human and environmental health. The emergence of interest in finding greener solvents to replace organic solvents has sparked a series of studies in the use of glycerol for extracting bioactive compounds from natural products. In this study, we will first identify the bioactive compounds of glycerol- and nonglycerol-based Thanaka (Hesperethusa crenulata) bark extracts using liquid chromatography-mass spectrometry profiles; then, we will determine their antioxidant capacity, free radical scavenging activity, and total phenolic and flavonoid contents. Thanaka bark powder was extracted using solvents, namely, ethanol (TKE), water (TKW), glycerol (TKG), glycerol/water (1:1, v/v) (TKGW), and glycerol/ethanol (1:1, v/v) (TKGE). Among the five extracts, the extract of TKG has the highest number of bioactive compounds, as well as the highest total flavonoid content. TKGE possessed the highest total phenolic content and highest antioxidant activity shown in azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) and ferric-reducing antioxidant power assays among the five extracts. Overall, glycerol has better efficiency in extracting bioactive compounds from Thanaka bark as compared to ethanol and water. Hence, from the phytochemical content and antioxidant properties of Thanaka extracts, we conclude that glycerol is a good green solvent alternative to replace organic solvents.
Collapse
Affiliation(s)
- Min Wen Lim
- Department
of Biological Sciences, School of Medical and Life Sciences, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Yin Quan Tang
- School
of Biosciences, Faculty of Health & Medical Sciences, Taylor’s University, No. 1, Jalan Taylors, 47500 Subang Jaya, Selangor, Malaysia
| | - Mohamed Kheireddine Aroua
- Centre
for Carbon Dioxide Capture and Utilization (CCDCU), School of Engineering
and Technology, Sunway University, No. 5, Jalan Universiti, Bandar
Sunway, 47500 Petaling
Jaya, Malaysia
- School
of Engineering, Lancaster University, LA1 4YW Lancaster, U.K.
| | - Lai Ti Gew
- Department
of Biological Sciences, School of Medical and Life Sciences, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| |
Collapse
|
8
|
Chen X, An Y, Tan M, Xie D, Liu L, Xu B. Biological functions and research progress of eIF4E. Front Oncol 2023; 13:1076855. [PMID: 37601696 PMCID: PMC10435865 DOI: 10.3389/fonc.2023.1076855] [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: 11/04/2022] [Accepted: 01/30/2023] [Indexed: 08/22/2023] Open
Abstract
The eukaryotic translation initiation factor eIF4E can specifically bind to the cap structure of an mRNA 5' end, mainly regulating translation initiation and preferentially enhancing the translation of carcinogenesis related mRNAs. The expression of eIF4E is closely related to a variety of malignant tumors. In tumor cells, eIF4E activity is abnormally increased, which stimulates cell growth, metastasis and translation of related proteins. The main factors affecting eIF4E activity include intranuclear regulation, phosphorylation of 4EBPs, and phosphorylation and sumoylation of eIF4E. In this review, we summarize the biological functions and the research progress of eIF4E, the main influencing factors of eIF4E activity, and the recent progress of drugs targeting eIF4E, in the hope of providing new insights for the treatment of multiple malignancies and development of targeted drugs.
Collapse
Affiliation(s)
- Xiaocong Chen
- Department of Clinical Medicine, Fenyang College of Shanxi Medical University, Fenyang, China
| | - Yang An
- Department of Clinical Medicine, Fenyang College of Shanxi Medical University, Fenyang, China
| | - Mengsi Tan
- Department of Clinical Medicine, Fenyang College of Shanxi Medical University, Fenyang, China
| | - Dongrui Xie
- Department of Clinical Medicine, Fenyang College of Shanxi Medical University, Fenyang, China
| | - Ling Liu
- Department of Medical Laboratory Science, Fenyang College of Shanxi Medical University, Fenyang, China
- Key Laboratory of Lvliang for Clinical Molecular Diagnostics, Fenyang, China
- Department of Clinical Laboratory, Fenyang Hospital of Shanxi Province, Fenyang, China
| | - Benjin Xu
- Department of Medical Laboratory Science, Fenyang College of Shanxi Medical University, Fenyang, China
- Key Laboratory of Lvliang for Clinical Molecular Diagnostics, Fenyang, China
- Department of Clinical Laboratory, Fenyang Hospital of Shanxi Province, Fenyang, China
| |
Collapse
|
9
|
Piteša N, Kurtović M, Bartoniček N, Gkotsi DS, Čonkaš J, Petrić T, Musani V, Ozretić P, Riobo-Del Galdo NA, Sabol M. Signaling Switching from Hedgehog-GLI to MAPK Signaling Potentially Serves as a Compensatory Mechanism in Melanoma Cell Lines Resistant to GANT-61. Biomedicines 2023; 11:1353. [PMID: 37239024 PMCID: PMC10216463 DOI: 10.3390/biomedicines11051353] [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: 03/22/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Melanoma represents the deadliest skin cancer due to its cell plasticity which results in high metastatic potential and chemoresistance. Melanomas frequently develop resistance to targeted therapy; therefore, new combination therapy strategies are required. Non-canonical signaling interactions between HH-GLI and RAS/RAF/ERK signaling were identified as one of the drivers of melanoma pathogenesis. Therefore, we decided to investigate the importance of these non-canonical interactions in chemoresistance, and examine the potential for HH-GLI and RAS/RAF/ERK combined therapy. METHODS We established two melanoma cell lines resistant to the GLI inhibitor, GANT-61, and characterized their response to other HH-GLI and RAS/RAF/ERK inhibitors. RESULTS We successfully established two melanoma cell lines resistant to GANT-61. Both cell lines showed HH-GLI signaling downregulation and increased invasive cell properties like migration potential, colony forming capacity, and EMT. However, they differed in MAPK signaling activity, cell cycle regulation, and primary cilia formation, suggesting different potential mechanisms responsible for resistance occurrence. CONCLUSIONS Our study provides the first ever insights into cell lines resistant to GANT-61 and shows potential mechanisms connected to HH-GLI and MAPK signaling which may represent new hot spots for noncanonical signaling interactions.
Collapse
Affiliation(s)
- Nikolina Piteša
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Matea Kurtović
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Nenad Bartoniček
- The Garvan Institute of Medical Research, Genome Informatics, Genomics & Epigenetics Division, 384 Victoria St., Darlinghurst, NSW 2010, Australia;
- The Kinghorn Centre for Clinical Genomics, 370 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Danai S. Gkotsi
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; (D.S.G.); (N.A.R.-D.G.)
- Astbury Centre for Molecular Structural Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Josipa Čonkaš
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Tina Petrić
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Vesna Musani
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Petar Ozretić
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Natalia A. Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; (D.S.G.); (N.A.R.-D.G.)
- Astbury Centre for Molecular Structural Biology, University of Leeds, Leeds LS2 9JT, UK
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK
- Leeds Cancer Research Centre, University of Leeds, Leeds LS2 9JT, UK
| | - Maja Sabol
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| |
Collapse
|
10
|
Edamana S, Pedersen SF, Nejsum LN. Aquaporin water channels affect the response of conventional anticancer therapies of 3D grown breast cancer cells. Biochem Biophys Res Commun 2023; 639:126-133. [PMID: 36481356 DOI: 10.1016/j.bbrc.2022.11.096] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022]
Abstract
Aquaporin (AQP) water channels facilitate water transport across cellular membranes and are essential in regulation of body water balance. Moreover, several AQPs are overexpressed or ectopically expressed in breast cancer. Interestingly, several in vitro studies have suggested that AQPs can affect the response to conventional anticancer chemotherapies. Therefore, we took a systematic approach to test how AQP1, AQP3 and AQP5, which are often over-/ectopically expressed in breast cancer, affect total viability of 3-dimensional (3D) breast cancer cell spheroids when treated with the conventional anticancer chemotherapies Cisplatin, 5-Fluorouracil (5-FU) and Doxorubicin, a Combination of the three drugs as well as the Combination plus the Ras inhibitor Salirasib. Total viability of spheroids overexpressing AQP1 were decreased by all treatments except for 5-FU, which increased total viability by 20% compared to DMSO treated controls. All treatments reduced viability of spheroids overexpressing AQP3. In contrast, only Doxorubicin, Combination and Combination + Salirasib reduced total viability of spheroids overexpressing AQP5. Thus, this study supports a significant role of AQPs in the response to conventional chemotherapies. Evaluating the role of individual proteins that contribute to resistance to chemotherapies is essential in advancing personalized medicine in breast carcinomas.
Collapse
Affiliation(s)
- Sarannya Edamana
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark
| | - Stine F Pedersen
- Department of Biology, Section for Cell Biology and Physiology, University of Copenhagen, Universitetsparken 13, 2100, København Ø, Denmark
| | - Lene N Nejsum
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark.
| |
Collapse
|
11
|
Morstein J, Bader T, Cardillo AL, Schackmann J, Ashok S, Hougland JL, Hrycyna CA, Trauner DH, Distefano MD. Photoswitchable Isoprenoid Lipids Enable Optical Control of Peptide Lipidation. ACS Chem Biol 2022; 17:2945-2953. [PMID: 36194691 PMCID: PMC9799063 DOI: 10.1021/acschembio.2c00645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Photoswitchable lipids have emerged as attractive tools for the optical control of lipid bioactivity, metabolism, and biophysical properties. Their design is typically based on the incorporation of an azobenzene photoswitch into the hydrophobic lipid tail, which can be switched between its trans- and cis-form using two different wavelengths of light. While glycero- and sphingolipids have been successfully designed to be photoswitchable, isoprenoid lipids have not yet been investigated. Herein, we describe the development of photoswitchable analogs of an isoprenoid lipid and systematically assess their potential for the optical control of various steps in the isoprenylation processing pathway of CaaX proteins in Saccharomyces cerevisiae. One photoswitchable analog of farnesyl diphosphate (AzoFPP-1) allowed effective optical control of substrate prenylation by farnesyltransferase. The subsequent steps of isoprenylation processing (proteolysis by either Ste24 or Rce1 and carboxyl methylation by Ste14) were less affected by photoisomerization of the group introduced into the lipid moiety of the substrate a-factor, a mating pheromone from yeast. We assessed both proteolysis and methylation of the a-factor analogs in vitro and the bioactivity of a fully processed a-factor analog containing the photoswitch, exogenously added to cognate yeast cells. Combined, these data describe the first successful conversion of an isoprenoid lipid into a photolipid and suggest the utility of this approach for the optical control of protein prenylation.
Collapse
Affiliation(s)
- Johannes Morstein
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, California 94158, USA
- Department of Chemistry, New York University, New York, New York 10003, USA
| | - Taysir Bader
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Ariana L. Cardillo
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, USA
| | - Julian Schackmann
- Department of Chemistry, New York University, New York, New York 10003, USA
| | - Sudhat Ashok
- Department of Chemistry, Syracuse University, Syracuse, New York 13244, USA
| | - James L. Hougland
- Department of Chemistry, Syracuse University, Syracuse, New York 13244, USA
- Department of Biology, Syracuse University, Syracuse, New York 13244, USA
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, USA
| | | | - Dirk H. Trauner
- Department of Chemistry, New York University, New York, New York 10003, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Mark D. Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
12
|
Grazier JJ, Sylvester PW. Role of Galectins in Metastatic Breast Cancer. Breast Cancer 2022. [DOI: 10.36255/exon-publications-breast-cancer-galectins] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
13
|
Kaur N, Lum M, Lewis RE, Black AR, Black JD. A novel anti-proliferative PKCα-Ras-ERK signaling axis in intestinal epithelial cells. J Biol Chem 2022; 298:102121. [PMID: 35697074 PMCID: PMC9270260 DOI: 10.1016/j.jbc.2022.102121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/05/2022] [Accepted: 05/31/2022] [Indexed: 01/02/2023] Open
Abstract
We have previously shown that the serine/threonine kinase PKCα triggers MAPK/ERK kinase (MEK)-dependent G1→S cell cycle arrest in intestinal epithelial cells, characterized by downregulation of cyclin D1 and inhibitor of DNA-binding protein 1 (Id1) and upregulation of the cyclin-dependent kinase inhibitor p21Cip1. Here, we use pharmacological inhibitors, genetic approaches, siRNA-mediated knockdown, and immunoprecipitation to further characterize anti-proliferative ERK signaling in intestinal cells. We show that PKCα signaling intersects the Ras-Raf-MEK-ERK kinase cascade at the level of Ras small GTPases, and that anti-proliferative effects of PKCα require active Ras, Raf, MEK and ERK, core ERK pathway components that are also essential for pro-proliferative ERK signaling induced by epidermal growth factor (EGF). However, PKCα-induced anti-proliferative signaling differs from EGF signaling in that it is independent of the Ras guanine nucleotide exchange factors (Ras-GEFs), SOS1/2, and involves prolonged rather than transient ERK activation. PKCα forms complexes with A-Raf, B-Raf and C-Raf that dissociate upon pathway activation, and all three Raf isoforms can mediate PKCα-induced anti-proliferative effects. At least two PKCα-ERK pathways that collaborate to promote growth arrest were identified: one pathway requiring the Ras-GEF, RasGRP3, and H-Ras, leads to p21Cip1 upregulation, while additional pathway(s) mediate PKCα-induced cyclin D1 and Id1 downregulation. PKCα also induces ERK-dependent SOS1 phosphorylation, indicating possible negative crosstalk between anti-proliferative and growth-promoting ERK signaling. Importantly, the spatio-temporal activation of PKCα and ERK in the intestinal epithelium in vivo supports the physiological relevance of these pathways and highlights the importance of anti-proliferative ERK signaling to tissue homeostasis in the intestine.
Collapse
Affiliation(s)
- Navneet Kaur
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Michelle Lum
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Robert E Lewis
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Adrian R Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jennifer D Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
14
|
Živanović AS, Bukonjić AM, Jovanović-Stević S, Bogojeski J, Ćoćić D, Bijelić AP, Ratković ZR, Volarević V, Miloradović D, Tomović DL, Radić GP. Complexes of copper(II) with tetradentate S,O-ligands: Synthesis, characterization, DNA/albumin interactions, molecular docking simulations and antitumor activity. J Inorg Biochem 2022; 233:111861. [DOI: 10.1016/j.jinorgbio.2022.111861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/02/2022] [Accepted: 05/08/2022] [Indexed: 11/29/2022]
|
15
|
Schmukler E, Pinkas-Kramarski R. The interplay between Ras, Autophagy and cancer. ADVANCES IN CANCER BIOLOGY - METASTASIS 2021; 3:100014. [DOI: 10.1016/j.adcanc.2021.100014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
16
|
Dai YH, Chen GY, Tang CH, Huang WC, Yang JC, Wu YC. Drug Screening of Potential Multiple Target Inhibitors for Estrogen Receptor-α-positive Breast Cancer. In Vivo 2021; 35:761-777. [PMID: 33622869 DOI: 10.21873/invivo.12317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM Estrogen receptor α (ERα) antagonist is the most common treatment for ERα-positive breast cancer. However, compensatory signaling contributes to resistance to ERα antagonists. Thus, to explore the potential agents for targeting compensatory signaling, we screened multiple target inhibitors for breast cancer treatment. MATERIALS AND METHODS We attempted to build a structure-based virtual screening model that can find potential compounds and assay the anticancer ability of these drugs by overall cell survival assay. The downstream compensatory phosphorylated signaling was measured by immunoblotting. RESULTS Hamamelitannin and glucocheirolin were hits for ERα, phosphoinositide 3-kinase (PI3K), and KRAS proto-oncogene, GTPase (KRAS), which were active against estrogen and epidermal growth factor-triggered proliferation. Additionally, we select aminopterin as a hit for ERα, PI3K, KRAS, and SRC proto-oncogene, non-receptor tyrosine kinase (SRC) with inhibitory activities toward AKT serine/threonine kinase 1 (AKT) and mitogen-activated protein kinase kinase (MEK) signaling. CONCLUSION Our structure-based virtual screening model selected hamamelitannin, glucocheirolin, aminopterin, and pemetrexed as compounds that may act as potential inhibitors for improving endocrine therapies for breast cancer.
Collapse
Affiliation(s)
- Yun-Hao Dai
- School of Pharmacy, China Medical University, Taichung, Taiwan, R.O.C.,Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C
| | - Guan-Yu Chen
- Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan, R.O.C.,Chinese Medicine Research Center, Drug Development Center, China Medical University, Taichung, Taiwan, R.O.C.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan, R.O.C
| | - Wei-Chien Huang
- Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C.,Chinese Medicine Research Center, Drug Development Center, China Medical University, Taichung, Taiwan, R.O.C.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,The Biotechnology Department, College of Medical and Health Science, Asia University, Taichung, Taiwan, R.O.C
| | - Juan-Cheng Yang
- Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C.;
| | - Yang-Chang Wu
- Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C.; .,The Biotechnology Department, College of Medical and Health Science, Asia University, Taichung, Taiwan, R.O.C.,Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan, R.O.C
| |
Collapse
|
17
|
Wang T, Mao P, Feng Y, Cui B, Zhang B, Chen C, Xu M, Gao K. Blocking hsa_circ_0006168 suppresses cell proliferation and motility of human glioblastoma cells by regulating hsa_circ_0006168/miR-628-5p/IGF1R ceRNA axis. Cell Cycle 2021; 20:1181-1194. [PMID: 34024251 PMCID: PMC8265815 DOI: 10.1080/15384101.2021.1930357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/30/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND hsa_circ_0006168 is an oncogenic circular RNA in esophageal cancer. However, its role remains unclarified in tumor progression of gliomas, especially in glioblastoma (GBM). METHODS Cell counting kit-8 assay, transwell assays, western blotting, and xenograft experiment, as well as colony formation assay and flow cytometry were performed to measure cell proliferation and motility. Expression of hsa_circ_0006168, microRNA (miR)-628-3p, insulin‑like growth factor 1 receptor (IGF1R), and Ras/extracellular signal regulated kinases (Erk)-related proteins were determined by quantitative real-time polymerase chain reaction and western blotting. The physical interaction was confirmed by dual-luciferase reporter assay and RNA pull-down assay. RESULTS hsa_circ_0006168 and IGF1R were upregulated, and miR-628-5p was downregulated in human GBM tissues and cells. Functionally, blocking hsa_circ_0006168 and overexpressing miR-628-5p suppressed cell proliferation, migration, invasion, and expression of Vimentin and Snail (mesenchymal markers) in A172 and LN229 cells, accompanied with increased E-cadherin (epithelial marker), decreased colony formation, and promoted apoptosis rate. Silencing miR-628-5p counteracted the suppression of hsa_circ_0006168 deficiency on these behaviors, and restoring IGF1R blocked miR-628-5p-mediated inhibition as well. More importantly, hsa_circ_0006168 knockdown could delay xenograft tumor growth in vivo and lower Ras and phosphorylated Erk1/2 expression in vitro and in vivo. Mechanically, hsa_circ_0006168 targeted and sponged miR-628-5p, and IFG1R was a novel target for miR-628-5p. Inhibiting miR-628-5p could abrogate in vitro role of hsa_circ_0006168 knockdown, and similarly IGF1R upregulation counteracted miR-628-5p role. CONCLUSION Silencing hsa_circ_0006168 might suppress GBM proliferation and motility via serving as competitive endogenous RNA for miR-628-5p and regulating IGF1R/Ras/Erk pathway.
Collapse
Affiliation(s)
- Tuo Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi, China
| | - Ping Mao
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi, China
| | - Yong Feng
- Department of Neurosurgery, The Hancheng People's Hospital, Weinan, Shannxi, China
| | - Bo Cui
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi, China
| | - Bin Zhang
- Department of Neurosurgery, Bao Ji Affiliated Hospital of Xi'an Medical University, Baoji, Shannxi, China
| | - Chen Chen
- Department of Neurosurgery, Mianxian Hospital, Mianxian, Shannxi, China
| | - Mingjie Xu
- Department of Neurosurgery, Traditional Chinese Medicine Hospital of Xixiang, Hanzhong, Shannxi, China
| | - Ke Gao
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi, China
| |
Collapse
|
18
|
Du R, Sullivan DK, Azizian NG, Liu Y, Li Y. Inhibition of ERAD synergizes with FTS to eradicate pancreatic cancer cells. BMC Cancer 2021; 21:237. [PMID: 33676427 PMCID: PMC7937230 DOI: 10.1186/s12885-021-07967-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC), one of the most lethal cancers, is driven by oncogenic KRAS mutations. Farnesyl thiosalicylic acid (FTS), also known as salirasib, is a RAS inhibitor that selectively dislodges active RAS proteins from cell membrane, inhibiting downstream signaling. FTS has demonstrated limited therapeutic efficacy in PDAC patients despite being well tolerated. Methods To improve the efficacy of FTS in PDAC, we performed a genome-wide CRISPR synthetic lethality screen to identify genetic targets that synergize with FTS treatment. Among the top candidates, multiple genes in the endoplasmic reticulum-associated protein degradation (ERAD) pathway were identified. The role of ERAD inhibition in enhancing the therapeutic efficacy of FTS was further investigated in pancreatic cancer cells using pharmaceutical and genetic approaches. Results In murine and human PDAC cells, FTS induced unfolded protein response (UPR), which was further augmented upon treatment with a chemical inhibitor of ERAD, Eeyarestatin I (EerI). Combined treatment with FTS and EerI significantly upregulated the expression of UPR marker genes and induced apoptosis in pancreatic cancer cells. Furthermore, CRISPR-based genetic ablation of the key ERAD components, HRD1 and SEL1L, sensitized PDAC cells to FTS treatment. Conclusion Our study reveals a critical role for ERAD in therapeutic response of FTS and points to the modulation of UPR as a novel approach to improve the efficacy of FTS in PDAC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07967-6.
Collapse
Affiliation(s)
- Rong Du
- Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.,Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Delaney K Sullivan
- UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Nancy G Azizian
- Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Yuanhui Liu
- Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Yulin Li
- Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
19
|
Baby B, Devan AR, Nair B, Nath LR. The Impetus of COVID -19 in Multiple Organ Affliction Apart from Respiratory Infection: Pathogenesis, Diagnostic Measures and Current Treatment Strategy. Infect Disord Drug Targets 2021; 21:514-526. [PMID: 32888278 DOI: 10.2174/1871526520999200905115050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 02/08/2023]
Abstract
The pandemic spread of COVID 19 caused by the novel Coronavirus (SARS-CoV- 2) produced a tremendous effect on the life of humanity across the globe. The epidemiological studies revealed the drastic spectrum of SARS-CoV 2 infection ranging from mere flu-like symptoms to severe respiratory suppression within a short period. Initially, cases have been confined in the emerging point, Wuhan, China. But, within a few months, it has spread all over 212 countries around the globe and presently has become a severe threat to human life. Even though it is a severe acute respiratory syndrome virus, recent reports came with multiple organ effects of SARS-CoV 2, suggesting the virulence potential of this novel virus to sweep the planet in the absence of a proper vaccine or therapy. In this review, we discuss the multi-organ pathophysiology of COVID-19 infection, together with the treatment methods adopted and innovative diagnostic methods used.
Collapse
Affiliation(s)
- Bilha Baby
- Amrita School of Pharmacy, Amrita VishwaVidyapeetham, AIMS Health Sciences Campus, AIMS P. O. Ponekkara, Kochi, Kerala-682041, India
| | - Aswathy R Devan
- Amrita School of Pharmacy, Amrita VishwaVidyapeetham, AIMS Health Sciences Campus, AIMS P. O. Ponekkara, Kochi, Kerala-682041, India
| | - Bhagyalakshmi Nair
- Amrita School of Pharmacy, Amrita VishwaVidyapeetham, AIMS Health Sciences Campus, AIMS P. O. Ponekkara, Kochi, Kerala-682041, India
| | - Lekshmi R Nath
- Amrita School of Pharmacy, Amrita VishwaVidyapeetham, AIMS Health Sciences Campus, AIMS P. O. Ponekkara, Kochi, Kerala-682041, India
| |
Collapse
|
20
|
Abstract
Ras proteins mediate extracellular and cytoplasmic signaling networks via receptor tyrosine kinase. The Ras pathway induces activation of signaling molecules involved in cell proliferation and growth, cell survival and apoptosis, metabolism, and motility. Although Ras mutations in breast cancer are not frequently reported, hyperactivation of Ras signaling plays an important role in breast cancer growth and progression. Oncogenic Ras activation occurs via loss of Ras GTPase-activating proteins, overexpression of growth factor receptor, and stimulation by various cytokines. Effective control of oncogenic Ras is one of the therapeutic strategies in breast cancer. The mechanisms of intracellular localization, activation, and signaling pathway of Ras in cancer have been used to develop therapeutic candidates. Recent studies have reported an effective therapy for breast cancer by inhibition of enzymes involved in the posttranslational modification of Ras, such as farnesyltransferase and geranylgeranyltransferase 1, and anti-cancer therapies targeting the epidermal growth factor receptor (EGFR). Emerging targets involved in EGF-mediated Ras activity in breast cancer have shed new insight into Ras activation in breast cancer progression. These alternative mechanisms for Ras signaling pathway may suggest novel therapeutic approaches for targeting Ras in breast cancer. In spite of the difficulties in targeting Ras protein, important discoveries highlight the direct inhibition of Ras activity. Further studies may elucidate the effects of targeting Ras protein and the clinical relevance thereof.
Collapse
|
21
|
Nagy Á, Győrffy B. muTarget: A platform linking gene expression changes and mutation status in solid tumors. Int J Cancer 2020; 148:502-511. [PMID: 32875562 DOI: 10.1002/ijc.33283] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/22/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
Large oncology repositories have paired genomic and transcriptomic data for all patients. We used these data to perform two independent analyses: to identify gene expression changes related to a gene mutation and to identify mutations altering the expression of a selected gene. All data processing steps were performed in the R statistical environment. RNA-sequencing and mutation data were acquired from The Cancer Genome Atlas (TCGA). The DESeq2 algorithm was applied for RNA-seq normalization, and transcript variants were annotated with AnnotationDbi. MuTect2-identified somatic mutation data were utilized, and the MAFtools Bioconductor program was used to summarize the data. The Mann-Whitney U test was used for differential expression analysis. The established database contains 7876 solid tumors from 18 different tumor types with both somatic mutation and RNA-seq data. The utility of the approach is presented via three analyses in breast cancer: gene expression changes related to TP53 mutations, gene expression changes related to CDH1 mutations and mutations resulting in altered progesterone receptor (PGR) expression. The breast cancer database was split into equally sized training and test sets, and these data sets were analyzed independently. The highly significant overlap of the results (chi-square statistic = 16 719.7 and P < .00001) validates the presented pipeline. Finally, we set up a portal at http://www.mutarget.com enabling the rapid identification of novel mutational targets. By linking somatic mutations and gene expression, it is possible to identify biomarkers and potential therapeutic targets in different types of solid tumors. The registration-free online platform can increase the speed and reduce the development cost of novel personalized therapies.
Collapse
Affiliation(s)
- Ádám Nagy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary.,Momentum Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary.,Momentum Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary.,2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| |
Collapse
|
22
|
Abstract
RAS was identified as a human oncogene in the early 1980s and subsequently found to be mutated in nearly 30% of all human cancers. More importantly, RAS plays a central role in driving tumor development and maintenance. Despite decades of effort, there remain no FDA approved drugs that directly inhibit RAS. The prevalence of RAS mutations in cancer and the lack of effective anti-RAS therapies stem from RAS' core role in growth factor signaling, unique structural features, and biochemistry. However, recent advances have brought promising new drugs to clinical trials and shone a ray of hope in the field. Here, we will exposit the details of RAS biology that illustrate its key role in cell signaling and shed light on the difficulties in therapeutically targeting RAS. Furthermore, past and current efforts to develop RAS inhibitors will be discussed in depth.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States
| | - Imran Khan
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States
| | - John P O'Bryan
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States.
| |
Collapse
|
23
|
Uras IZ, Moll HP, Casanova E. Targeting KRAS Mutant Non-Small-Cell Lung Cancer: Past, Present and Future. Int J Mol Sci 2020; 21:E4325. [PMID: 32560574 PMCID: PMC7352653 DOI: 10.3390/ijms21124325] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the most frequent cancer with an aggressive clinical course and high mortality rates. Most cases are diagnosed at advanced stages when treatment options are limited and the efficacy of chemotherapy is poor. The disease has a complex and heterogeneous background with non-small-cell lung cancer (NSCLC) accounting for 85% of patients and lung adenocarcinoma being the most common histological subtype. Almost 30% of adenocarcinomas of the lung are driven by an activating Kirsten rat sarcoma viral oncogene homolog (KRAS) mutation. The ability to inhibit the oncogenic KRAS has been the holy grail of cancer research and the search for inhibitors is immensely ongoing as KRAS-mutated tumors are among the most aggressive and refractory to treatment. Therapeutic strategies tailored for KRAS+ NSCLC rely on the blockage of KRAS functional output, cellular dependencies, metabolic features, KRAS membrane associations, direct targeting of KRAS and immunotherapy. In this review, we provide an update on the most recent advances in anti-KRAS therapy for lung tumors with mechanistic insights into biological diversity and potential clinical implications.
Collapse
Affiliation(s)
- Iris Z. Uras
- Department of Pharmacology, Center of Physiology and Pharmacology & Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | - Herwig P. Moll
- Department of Physiology, Center of Physiology and Pharmacology & Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria; (H.P.M.); (E.C.)
| | - Emilio Casanova
- Department of Physiology, Center of Physiology and Pharmacology & Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria; (H.P.M.); (E.C.)
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), 1090 Vienna, Austria
| |
Collapse
|
24
|
Wang Y, Yang S, Zhang S, Wu X. Oxymatrine Inhibits Proliferation and Migration of Vulvar Squamous Cell Carcinoma Cells via Attenuation of the RAS/RAF/MEK/ERK Pathway. Cancer Manag Res 2020; 12:2057-2067. [PMID: 32256113 PMCID: PMC7090165 DOI: 10.2147/cmar.s245696] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/28/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose To evaluate the anti-tumor effects of oxymatrine in vulvar squamous cell carcinoma (VSCC) cells and to explore the underlying mechanisms. Methods We selected SW962 and A431 VSCC cell lines. Cell proliferation was examined using MTT assay. Cell cycle and apoptosis were detected using flow cytometry. Migration and invasion were evaluated using transwell and wound-healing assays. The relevant protein expression and signaling pathways were analyzed using Western blotting. Results Oxymatrine inhibited the proliferation of SW962 and A431 VSCC cells in a time- and dose-dependent manner. Oxymatrine induced cell cycle arrest in the G2/M phase by increasing the protein expression of P21 and decreasing levels of cyclin B1 and CDC2. Oxymatrine upregulated the expression of cleaved-caspase 3 and BAX and downregulated the expression of BCL2, which led to an increase in apoptosis. Oxymatrine also suppressed the migration and invasion of SW962 and A431 cells by reducing levels of MMP2 and MMP9. After treatment with oxymatrine or a RAS inhibitor (salirasib), expression levels of RAS, p-RAF, p-MEK, p-ERK, C-MYC, and MMP2 were reduced. When TGF-β1 was used to stimulate SW962 and A431 cells, the expression of the above proteins increased; this increase was reversed by using oxymatrine or salirasib again. Conclusion Oxymatrine inhibits proliferation and migration of VSCC cells by blocking the RAS/RAF/MEK/ERK pathway.
Collapse
Affiliation(s)
- Yanshi Wang
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| | - Shaojie Yang
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| | - Shanshan Zhang
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| | - Xin Wu
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| |
Collapse
|
25
|
Nasal Drug Delivery of Anticancer Drugs for the Treatment of Glioblastoma: Preclinical and Clinical Trials. Molecules 2019; 24:molecules24234312. [PMID: 31779126 PMCID: PMC6930669 DOI: 10.3390/molecules24234312] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/22/2019] [Accepted: 11/24/2019] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most lethal form of brain tumor, being characterized by the rapid growth and invasion of the surrounding tissue. The current standard treatment for glioblastoma is surgery, followed by radiotherapy and concurrent chemotherapy, typically with temozolomide. Although extensive research has been carried out over the past years to develop a more effective therapeutic strategy for the treatment of GBM, efforts have not provided major improvements in terms of the overall survival of patients. Consequently, new therapeutic approaches are urgently needed. Overcoming the blood–brain barrier (BBB) is a major challenge in the development of therapies for central nervous system (CNS) disorders. In this context, the intranasal route of drug administration has been proposed as a non-invasive alternative route for directly targeting the CNS. This route of drug administration bypasses the BBB and reduces the systemic side effects. Recently, several formulations have been developed for further enhancing nose-to-brain transport, mainly with the use of nano-sized and nanostructured drug delivery systems. The focus of this review is to provide an overview of the strategies that have been developed for delivering anticancer compounds for the treatment of GBM while using nasal administration. In particular, the specific properties of nanomedicines proposed for nose-to-brain delivery will be critically evaluated. The preclinical and clinical data considered supporting the idea that nasal delivery of anticancer drugs may represent a breakthrough advancement in the fight against GBM.
Collapse
|
26
|
Fan M, Xia P, Liu B, Zhang L, Wang Y, Gao X, Li L. Tumour heterogeneity revealed by unsupervised decomposition of dynamic contrast-enhanced magnetic resonance imaging is associated with underlying gene expression patterns and poor survival in breast cancer patients. Breast Cancer Res 2019; 21:112. [PMID: 31623683 PMCID: PMC6798414 DOI: 10.1186/s13058-019-1199-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 09/13/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Heterogeneity is a common finding within tumours. We evaluated the imaging features of tumours based on the decomposition of tumoural dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) data to identify their prognostic value for breast cancer survival and to explore their biological importance. METHODS Imaging features (n = 14), such as texture, histogram distribution and morphological features, were extracted to determine their associations with recurrence-free survival (RFS) in patients in the training cohort (n = 61) from The Cancer Imaging Archive (TCIA). The prognostic value of the features was evaluated in an independent dataset of 173 patients (i.e. the reproducibility cohort) from the TCIA I-SPY 1 TRIAL dataset. Radiogenomic analysis was performed in an additional cohort, the radiogenomic cohort (n = 87), using DCE-MRI from TCGA-BRCA and corresponding gene expression data from The Cancer Genome Atlas (TCGA). The MRI tumour area was decomposed by convex analysis of mixtures (CAM), resulting in 3 components that represent plasma input, fast-flow kinetics and slow-flow kinetics. The prognostic MRI features were associated with the gene expression module in which the pathway was analysed. Furthermore, a multigene signature for each prognostic imaging feature was built, and the prognostic value for RFS and overall survival (OS) was confirmed in an additional cohort from TCGA. RESULTS Three image features (i.e. the maximum probability from the precontrast MR series, the median value from the second postcontrast series and the overall tumour volume) were independently correlated with RFS (p values of 0.0018, 0.0036 and 0.0032, respectively). The maximum probability feature from the fast-flow kinetics subregion was also significantly associated with RFS and OS in the reproducibility cohort. Additionally, this feature had a high correlation with the gene expression module (r = 0.59), and the pathway analysis showed that Ras signalling, a breast cancer-related pathway, was significantly enriched (corrected p value = 0.0044). Gene signatures (n = 43) associated with the maximum probability feature were assessed for associations with RFS (p = 0.035) and OS (p = 0.027) in an independent dataset containing 1010 gene expression samples. Among the 43 gene signatures, Ras signalling was also significantly enriched. CONCLUSIONS Dynamic pattern deconvolution revealed that tumour heterogeneity was associated with poor survival and cancer-related pathways in breast cancer.
Collapse
Affiliation(s)
- Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Xiasha High Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Pingping Xia
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Xiasha High Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Bin Liu
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Xiasha High Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Lin Zhang
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Xiasha High Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| | - Xin Gao
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Xiasha High Education Zone, Hangzhou, 310018, Zhejiang, China.
| |
Collapse
|
27
|
He ST, Lee DY, Tung CY, Li CY, Wang HC. Glutamine Metabolism in Both the Oxidative and Reductive Directions Is Triggered in Shrimp Immune Cells (Hemocytes) at the WSSV Genome Replication Stage to Benefit Virus Replication. Front Immunol 2019; 10:2102. [PMID: 31555294 PMCID: PMC6737011 DOI: 10.3389/fimmu.2019.02102] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022] Open
Abstract
White spot syndrome virus (WSSV) is the causative agent of a shrimp disease that has caused huge global economic losses. Although its pathogenesis remains poorly understood, it has been reported that in the shrimp immune cells (hemocytes) targeted by WSSV, the virus triggers both the Warburg effect and glutamine metabolism at the WSSV genome replication stage (12 h post infection). Glutamine metabolism follows two pathways: an oxidative pathway mediated by α-KGDH (α-ketoglutarate dehydrogenase) and an alternative reductive pathway mediated by IDH1 and IDH2 (isocitrate dehydrogenase 1 and 2). Here we used isotopically labeled glutamine ([U-13C]glutamine and [1-13C]glutamine) as metabolic tracers to show that, at the replication stage, both the oxidative and reductive glutamine metabolic pathways were activated. We further show that the mRNA expression levels of α-KGDH and IDH1 were increased in WSSV-infected shrimps and that silencing of α-KGDH, IDH1, and IDH2 with their respective dsRNAs led to a decrease in WSSV gene expression and WSSV replication. Taken together, our findings provide new evidence for WSSV-induced metabolic reprogramming in hemocytes and demonstrate its importance in virus replication.
Collapse
Affiliation(s)
- Shu-Ting He
- Department of Biotechnology and Bioindustry Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Der-Yen Lee
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Yi Tung
- Department of Biotechnology and Bioindustry Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Yuan Li
- Department of Biotechnology and Bioindustry Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Han-Ching Wang
- Department of Biotechnology and Bioindustry Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, Taiwan.,International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
28
|
Lakkakula BVKS, Farran B, Lakkakula S, Peela S, Yarla NS, Bramhachari PV, Kamal MA, Saddala MS, Nagaraju GP. Small molecule tyrosine kinase inhibitors and pancreatic cancer-Trials and troubles. Semin Cancer Biol 2019; 56:149-167. [PMID: 30314681 DOI: 10.1016/j.semcancer.2018.09.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/18/2018] [Accepted: 09/29/2018] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer (PC) is an aggressive carcinoma and the fourth cause of cancer deaths in Western countries. Although surgery is the most effective therapeutic option for PC, the management of unresectable, locally advanced disease is highly challenging. Our improved understanding of pancreatic tumor biology and associated pathways has led to the development of various treatment modalities that can control the metastatic spread of PC. This review intends to present trials of small molecule tyrosine kinase inhibitors (TKIs) in PC management and the troubles encountered due to inevitable acquired resistance to TKIs.
Collapse
Affiliation(s)
| | - Batoul Farran
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA-30322, USA
| | - Saikrishna Lakkakula
- Department of Zoology, Visvodaya Government Degree College, Venkatagiri, AP-524132, India
| | - Sujatha Peela
- Department of Biotechnology, Dr.B.R.Ambedkar University, Srikakulam, Andhra Pradesh, India
| | - Nagendra Sastry Yarla
- Dr. LV Prasad Diagnostics and Research Laboratory, Khairtabad, Hyderabad, AP- 500004, India
| | | | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Australia; Novel Global Community Educational Foundation, Australia
| | | | - Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA-30322, USA.
| |
Collapse
|
29
|
Tseng YT, Kumar R, Wang HC. LvRas and LvRap are both important for WSSV replication in Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2019; 88:150-160. [PMID: 30794934 DOI: 10.1016/j.fsi.2019.02.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 06/09/2023]
Abstract
The white Spot Syndrome Virus (WSSV) is a pathogen that causes huge economic losses in the shrimp-farming industry globally. At the WSSV genome replication stage (12 hpi) in WSSV-infected shrimp hemocytes, activation of the PI3K-Akt-mTOR pathway triggers metabolic changes that resemble the Warburg effect. In shrimp, the upstream regulators of this pathway are still unknown, and in the present study, we isolate, characterize and investigate two candidate factors, i.e. the shrimp Ras GTPase isoforms LvRas and LvRap, both of which are upregulated after WSSV infection. dsRNA silencing experiments show that virus replication is significantly reduced when expression of either of these genes is suppressed. Pretreatment with the Ras inhibitor Salirasib further suggests that LvRas, which is a homolog to a commonly overexpressed human oncoprotein, may be involved in regulating the WSSV-induced Warburg effect. We also show that while both the PI3K-Akt-mTOR and Raf-MEK-ERK pathways are activated by WSSV infection, LvRas appears to be involved only in the regulation of the mTOR pathway.
Collapse
Affiliation(s)
- Yi-Ting Tseng
- Department of Biotechnology and Bioindustry Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Ramya Kumar
- Department of Biotechnology and Bioindustry Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Han-Ching Wang
- Department of Biotechnology and Bioindustry Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan; International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
30
|
Xu A, Wang B, Fu J, Qin W, Yu T, Yang Z, Lu Q, Chen J, Chen Y, Wang H. Diet-induced hepatic steatosis activates Ras to promote hepatocarcinogenesis via CPT1α. Cancer Lett 2018; 442:40-52. [PMID: 30401637 DOI: 10.1016/j.canlet.2018.10.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/06/2018] [Accepted: 10/17/2018] [Indexed: 01/22/2023]
Abstract
Aberrant activation of the RAS cascade ubiquitously occurs in human hepatocellular carcinomas (HCC), regardless of rare mutations of RAS. However, the association between the Ras cascade and hepatic steatosis during hepatocarcinogenesis remains under-investigated. Here, the variation in the constitutive activity of Ras signaling and HCC incidence was found in a nonalcoholic fatty liver disease (NAFLD)-HCC mouse model, and Ras activity was induced by hepatic steatosis. Even in hepatocyte-specific expression of KrasG12D (Alb-Cre/KrasG12D, Krashep) mice, mutagenic activation of Ras signaling was still significantly enhanced by NAFLD, with downregulation of negative regulators. Interestingly, hepatic steatosis could be alleviated by persistent activation of Ras, whereas Ras accelerated DNA damage and HCC progression through Carnitine palmitoyltransferase 1A (CPT1α). A close correlation between active Ras and CPT1α was also shown in clinical steatosis peri-tumor tissues of HCC samples and experimental models. CPT1α inhibitor etomoxir (ETO) largely ameliorated active Ras-drived HCC. These findings can provide a novel link between steatosis and Ras activity in liver cancer.
Collapse
Affiliation(s)
- An Xu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Bibo Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China; Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jing Fu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Wenhao Qin
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Ting Yu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China; Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Zhishi Yang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Qingjun Lu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyi Chen
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Yao Chen
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China.
| | - Hongyang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China; Fuling Central Hospital of Chongqing City, Chongqing, China.
| |
Collapse
|
31
|
Sun J, Sun L, Li J, Xu J, Wan Z, Ouyang Z, Liang L, Li S, Zeng D. A multi-functional polymeric carrier for simultaneous positron emission tomography imaging and combination therapy. Acta Biomater 2018; 75:312-322. [PMID: 29885530 PMCID: PMC6119490 DOI: 10.1016/j.actbio.2018.06.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/17/2018] [Accepted: 06/05/2018] [Indexed: 01/06/2023]
Abstract
Multifunctional nanoplatforms offering simultaneous imaging and therapeutic functions have been recognized as a highly promising strategy for personalized nanomedicine. In this work, we synthesized a farnesylthiosalicylate (FTS, a nontoxic Ras antagonist) based triblock copolymer POEG-b-PVBA-b-PFTS (POVF) composed of a poly(oligo(ethylene glycol) methacrylate) (POEG) hydrophilic block, a poly(FTS) hydrophobic block, and a poly(4-vinylbenzyl azide) (PVBA) middle block. The POVF polymer itself was active in inhibiting the tumor growth in vitro and in vivo. Besides, it could serve as a carrier to effectively encapsulate paclitaxel (PTX) to form stable PTX/POVF mixed micelles with a diameter around 100 nm. Meanwhile, POVF polymer provides the active azide group for incorporating a positron emission tomography (PET) imaging modality via a facile strategy based on metal-free click chemistry. This nanocarrier system could not only be used for co-delivery of PTX and FTS, but also for PET imaging guided drug delivery. In the 4T1.2 tumor bearing mice, PET imaging showed rapid uptake and slow clearance of radiolabeled PTX/POVF nanomicelles in the tumor tissues. In addition, the FTS-based multi-functional nanocarrier was able to inhibit tumor growth effectively, and the co-delivery of PTX by the carrier further improved the therapeutic effect. STATEMENT OF SIGNIFICANCE Due to the intrinsic heterogeneity of cancer and variability in individual patient response, personalized nanomedicine based on multi-functional carriers that integrate the functionalities of combination therapy and imaging guidance is highly demanded. Here we developed a multi-functional nanocarrier based on triblock copolymer POEG-b-PVBA-b-PFTS (POVF), which could not only be used for co-delivery of anticancer drugs PTX and Ras inhibitor FTS, but also for PET imaging guided drug delivery. The POVF carrier itself was active in inhibiting the tumor growth in vitro and in vivo. Besides, it was effective in formulating PTX with high drug loading capacity, which further enhanced the tumor inhibition effect. Meanwhile, we developed a simple and universal approach to incorporate a PET radioisotope (Zr-89 and Cu-64) into the azide-containing PTX/POVF micelles via metal-free click chemistry in aqueous solution. The radiolabeled PTX/POVF micelles exhibited excellent serum stability, rapid tumor uptake and slow clearance, which validated the feasibility of the PET image-guided delivery of PTX/POVF micelles.
Collapse
Affiliation(s)
- Jingjing Sun
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Lingyi Sun
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Jianchun Li
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Jieni Xu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Zhuoya Wan
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Zubin Ouyang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lei Liang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| | - Dexing Zeng
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, United States.
| |
Collapse
|
32
|
Husein A, Jamal A, Ahmed MZ, Arish M, Ali R, Tabrez S, Rasool F, Rub A. Leishmania donovani infection differentially regulates small G-proteins. J Cell Biochem 2018; 119:7844-7854. [PMID: 29943842 DOI: 10.1002/jcb.27186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 05/24/2018] [Indexed: 01/28/2023]
Abstract
Leishmania is a protozoan parasite that resides and replicates in macrophages and causes leishmaniasis. The parasite alters the signaling cascade in host macrophages and evades the host machinery. Small G-proteins are GTPases, grouped in 5 different families that play a crucial role in the regulation of cell proliferation, cell survival, apoptosis, intracellular trafficking, and transport. In particular, the Ras family of small G-proteins has been identified to play a significant role in the cellular functions mentioned before. Here, we studied the differential expression of the most important small G-proteins during Leishmania infection. We found major changes in the expression of different isoforms of Ras, mainly in N-Ras. We observed that Leishmania donovani infection led to enhanced N-Ras expression, whereas it inhibited K-Ras and H-Ras expression. Furthermore, an active N-Ras pull-down assay showed enhanced N-Ras activity. L donovani infection also increased extracellular signal-regulated kinase 1/2 phosphorylation and simultaneously decreased p38 phosphorylation. In contrast, pharmacological inhibition of Ras led to reduction in the phosphorylation of extracellular signal-regulated kinase 1/2 and enhanced the phosphorylation of p38 in Leishmania-infected cells, which could lead to increased interleukin-12 expression and decreased interleukin-10 expression. Indeed, farnesylthiosalicyclic acid (a Ras inhibitor), when used at the effective level in L donovani-infected macrophages, reduced amastigotes in the host macrophages. Thus, upregulated N-Ras expression during L donovani infection could be a novel immune evasion strategy of Leishmania and would be a potential target for antileishmanial immunotherapy.
Collapse
Affiliation(s)
- Atahar Husein
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Azfar Jamal
- Virology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Mohammad Zulfazal Ahmed
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Arish
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Rahat Ali
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Shams Tabrez
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Fayyaz Rasool
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Abdur Rub
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.,Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| |
Collapse
|
33
|
Mallangada NA, Vargas JM, Thomas S, DiGiovanni MG, Vaeth BM, Nemesure MD, Wang R, LaComb JF, Williams JL, Golub LM, Johnson F, Mackenzie GG. A novel tricarbonylmethane agent (CMC2.24) reduces human pancreatic tumor growth in mice by targeting Ras. Mol Carcinog 2018; 57:1130-1143. [PMID: 29683208 DOI: 10.1002/mc.22830] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/24/2018] [Accepted: 04/17/2018] [Indexed: 12/28/2022]
Abstract
Pancreatic Cancer (PC) is a deadly disease in need of new therapeutic options. We recently developed a novel tricarbonylmethane agent (CMC2.24) as a therapeutic agent for PC, and evaluated its efficacy in preclinical models of PC. CMC2.24 inhibited the growth of various human PC cell lines in a concentration and time-dependent manner. Normal human pancreatic epithelial cells were resistant to CMC2.24, indicating selectivity. CMC2.24 reduced the growth of subcutaneous and orthotopic PC xenografts in mice by up to 65% (P < 0.02), and the growth of a human patient-derived tumor xenograft by 47.5% (P < 0.03 vs vehicle control). Mechanistically, CMC2.24 inhibited the Ras-RAF-MEK-ERK pathway. Based on Ras Pull-Down Assays, CMC2.24 inhibited Ras-GTP, the active form of Ras, in MIA PaCa-2 cells and in pancreatic acinar explants isolated from Kras mutant mice, by 90.3% and 89.1%, respectively (P < 0.01, for both). The inhibition of active Ras led to an inhibition of c-RAF, MEK, and ERK phosphorylation by 93%, 91%, and 87%, respectively (P < 0.02, for all) in PC xenografts. Furthermore, c-RAF overexpression partially rescued MIA PaCa-2 cells from the cell growth inhibition by CMC2.24. In addition, downstream of ERK, CMC2.24 inhibited STAT3 phosphorylation levels at the serine 727 residue, enhanced the levels of superoxide anion in mitochondria, and induced intrinsic apoptosis as shown by the release of cytochrome c from the mitochondria to the cytosol and the further cleavage of caspase 9 in PC cells. In conclusion, CMC2.24, a potential Ras inhibitor, is an efficacious agent for PC treatment in preclinical models, deserving further evaluation.
Collapse
Affiliation(s)
- Naveen A Mallangada
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Joselin M Vargas
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Swaroopa Thomas
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Matthew G DiGiovanni
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Brandon M Vaeth
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Matthew D Nemesure
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Ruixue Wang
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Joseph F LaComb
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Jennie L Williams
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Lorne M Golub
- Department of Oral Biology and Pathology, Stony Brook University, Stony Brook, New York
| | - Francis Johnson
- Departments of Chemistry and of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | - Gerardo G Mackenzie
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York.,Stony Brook Cancer Center, Stony Brook, New York.,Department of Nutrition, University of California, Davis, California
| |
Collapse
|
34
|
Lang L, Shay C, Zhao X, Teng Y. Combined targeting of Arf1 and Ras potentiates anticancer activity for prostate cancer therapeutics. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:112. [PMID: 28830537 PMCID: PMC5568197 DOI: 10.1186/s13046-017-0583-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023]
Abstract
Background Although major improvements have been made in surgical management, chemotherapeutic, and radiotherapeutic of prostate cancer, many prostate cancers remain refractory to treatment with standard agents. Therefore, the identification of new molecular targets in cancer progression and development of novel therapeutic strategies to target them are very necessary for achieving better survival for patients with prostate cancer. Activation of small GTPases such as Ras and Arf1 is a critical component of the signaling pathways for most of the receptors shown to be upregulated in advanced prostate cancer. Methods The drug effects on cell proliferation were measured by CellTiter 96® AQueous One Solution Cell Proliferation Assay. The drug effects on cell migration and invasion were determined by Radius™ 24-well and Matrigel-coated Boyden chambers. The drug effects on apoptosis were assessed by FITC Annexin V Apoptosis Detection Kit with 7-AAD and Western blot with antibodies against cleaved PARP and Caspase 3. A NOD/SCID mouse model generated by subcutaneous injection was used to assess the in vivo drug efficacy in tumor growth. ERK activation and tumor cell proliferation in xenografts were examined by immunohistochemistry. Results We show that Exo2, a small-molecule inhibitor that reduces Arf1 activation, effectively suppresses prostate cancer cell proliferation by blocking ERK1/2 activation. Exo2 also has other effects, inhibiting migration and invasion of PCa cells and inducing apoptosis. The Ras inhibitor salirasib augments Exo2-induced cytotoxicity in prostate cancer cells partially by enhancing the suppression of ERK1/2 phosphorylation. In a xenograft mouse model of prostate cancer, Exo2 reduces prostate tumor burden and inhibits ERK1/2 activation at a dose of 20 mg/kg. Synergistic treatment of salirasib and Exo2 exhibits a superior inhibitory effect on prostate tumor growth compared with either drug alone, which may be attributed to the more efficient inhibition of ERK1/2 phosphorylation. Conclusion This study suggests that simultaneous blockade of Arf1 and Ras activation in prostate cancer cells is a potential targeted therapeutic strategy for preventing prostate cancer development. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0583-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liwei Lang
- Department of Oral Biology, Augusta University, Augusta, GA, 30912, USA
| | - Chloe Shay
- Department of Pediatrics, Emory Children's Center, Emory University, Atlanta, GA, 30322, USA
| | - Xiangdong Zhao
- Department of Oral Biology, Augusta University, Augusta, GA, 30912, USA
| | - Yong Teng
- Department of Oral Biology, Augusta University, Augusta, GA, 30912, USA. .,Georgia Cancer Center, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA. .,Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
35
|
Engin HB, Carlin D, Pratt D, Carter H. Modeling of RAS complexes supports roles in cancer for less studied partners. BMC BIOPHYSICS 2017; 10:5. [PMID: 28815022 PMCID: PMC5558186 DOI: 10.1186/s13628-017-0037-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background RAS protein interactions have predominantly been studied in the context of the RAF and PI3kinase oncogenic pathways. Structural modeling and X-ray crystallography have demonstrated that RAS isoforms bind to canonical downstream effector proteins in these pathways using the highly conserved switch I and II regions. Other non-canonical RAS protein interactions have been experimentally identified, however it is not clear whether these proteins also interact with RAS via the switch regions. Results To address this question we constructed a RAS isoform-specific protein-protein interaction network and predicted 3D complexes involving RAS isoforms and interaction partners to identify the most probable interaction interfaces. The resulting models correctly captured the binding interfaces for well-studied effectors, and additionally implicated residues in the allosteric and hyper-variable regions of RAS proteins as the predominant binding site for non-canonical effectors. Several partners binding to this new interface (SRC, LGALS1, RABGEF1, CALM and RARRES3) have been implicated as important regulators of oncogenic RAS signaling. We further used these models to investigate competitive binding and multi-protein complexes compatible with RAS surface occupancy and the putative effects of somatic mutations on RAS protein interactions. Conclusions We discuss our findings in the context of RAS localization to the plasma membrane versus within the cytoplasm and provide a list of RAS protein interactions with possible cancer-related consequences, which could help guide future therapeutic strategies to target RAS proteins. Electronic supplementary material The online version of this article (doi:10.1186/s13628-017-0037-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- H Billur Engin
- Division of Medical Genetics, Department of Medicine, Universsity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093 USA
| | - Daniel Carlin
- Division of Medical Genetics, Department of Medicine, Universsity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093 USA
| | - Dexter Pratt
- Division of Medical Genetics, Department of Medicine, Universsity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093 USA
| | - Hannah Carter
- Division of Medical Genetics, Department of Medicine, Universsity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093 USA
| |
Collapse
|
36
|
Li J, Ghazwani M, Liu K, Huang Y, Chang N, Fan J, He F, Li L, Bu S, Xie W, Ma X, Li S. Regulation of hepatic stellate cell proliferation and activation by glutamine metabolism. PLoS One 2017; 12:e0182679. [PMID: 28797105 PMCID: PMC5552314 DOI: 10.1371/journal.pone.0182679] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/21/2017] [Indexed: 12/20/2022] Open
Abstract
Liver fibrosis is the excessive accumulation of extracellular matrix proteins, which is mainly caused by accumulation of activated hepatic stellate cells (HSCs). The mechanisms of activation and proliferation of HSCs, two key events after liver damage, have been studied for many years. Here we report a novel pathway to control HSCs by regulating glutamine metabolism. We demonstrated that the proliferation of HSCs is critically dependent on glutamine that is used to generate α-ketoglutarate (α-KG) and non-essential amino acid (NEAA). In addition, both culture- and in vivo-activated HSCs have increased glutamine utilization and increased expression of genes related to glutamine metabolism, including GLS (glutaminase), aspartate transaminase (GOT1) and glutamate dehydrogenase (GLUD1). Inhibition of these enzymes, as well as glutamine depletion, had a significant inhibitory effect on HSCs activation. In addition to providing energy expenditure, conversion of glutamine to proline is enhanced. The pool of free proline may also be increased via downregulation of POX expression. Hedgehog signaling plays an important role in the regulation of glutamine metabolism, as well as TGF-β1, c-Myc, and Ras signalings, via transcriptional upregulation and repression of key metabolic enzymes in this pathway. Finally, changes in glutamine metabolism were also found in mouse liver tissue following CCl4-induced acute injury. CONCLUSION Glutamine metabolism plays an important role in regulating the proliferation and activation of HSCs. Strategies that are targeted at glutamine metabolism may represent a novel therapeutic approach to the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Jiang Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail: (JL); (SL)
| | - Mohammed Ghazwani
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ke Liu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Na Chang
- Department of Cell Biology,Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Jie Fan
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Fengtian He
- Department of Biochemistry and Molecular Biology (F.H.), College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Liying Li
- Department of Cell Biology,Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Shizhong Bu
- Diabetes Research Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail: (JL); (SL)
| |
Collapse
|
37
|
Pharmacological strategies to target oncogenic KRAS signaling in pancreatic cancer. Pharmacol Res 2017; 117:370-376. [DOI: 10.1016/j.phrs.2017.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 02/07/2023]
|
38
|
Schmukler E, Wolfson E, Elazar Z, Kloog Y, Pinkas-Kramarski R. Continuous treatment with FTS confers resistance to apoptosis and affects autophagy. PLoS One 2017; 12:e0171351. [PMID: 28151959 PMCID: PMC5289601 DOI: 10.1371/journal.pone.0171351] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/19/2017] [Indexed: 12/26/2022] Open
Abstract
High percentage of human cancers involves alteration or mutation in Ras proteins, including the most aggressive malignancies, such as lung, colon and pancreatic cancers. FTS (Salirasib) is a farnesylcysteine mimetic, which acts as a functional Ras inhibitor, and was shown to exert anti-tumorigenic effects in vitro and in vivo. Previously, we have demonstrated that short-term treatment with FTS also induces protective autophagy in several cancer cell lines. Drug resistance is frequently observed in cancer cells exposed to prolonged treatment, and is considered a major cause for therapy inefficiency. Therefore, in the present study, we examined the effect of a prolonged treatment with FTS on drug resistance of HCT-116 human colon cancer cells, and the involvement of autophagy in this process. We found that cells grown in the presence of FTS for 6 months have become resistant to FTS-induced cell growth inhibition and cell death. Furthermore, we discovered that the resistant cells exhibit altered autophagy, reduced apoptosis and changes in Ras-related signaling pathways following treatment with FTS. Moreover we found that while FTS induces an apoptosis-related cleavage of p62, the FTS-resistant cells were more resistant to apoptosis and p62 cleavage.
Collapse
Affiliation(s)
- Eran Schmukler
- Department of Neurobiology. Tel-Aviv University, Ramat-Aviv, Israel
| | - Eya Wolfson
- Department of Neurobiology. Tel-Aviv University, Ramat-Aviv, Israel
| | - Zvulun Elazar
- Department of Biological Chemistry; The Weizmann Institute of Science; Rehovot, Israel
| | - Yoel Kloog
- Department of Neurobiology. Tel-Aviv University, Ramat-Aviv, Israel
| | | |
Collapse
|
39
|
Tomasini P, Walia P, Labbe C, Jao K, Leighl NB. Targeting the KRAS Pathway in Non-Small Cell Lung Cancer. Oncologist 2016; 21:1450-1460. [PMID: 27807303 PMCID: PMC5153335 DOI: 10.1634/theoncologist.2015-0084] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/29/2016] [Indexed: 12/19/2022] Open
Abstract
: Lung cancer remains the leading cause of cancer-related deaths worldwide. However, significant progress has been made individualizing therapy based on molecular aberrations (e.g., EGFR, ALK) and pathologic subtype. KRAS is one of the most frequently mutated genes in non-small cell lung cancer (NSCLC), found in approximately 30% of lung adenocarcinomas, and is thus an appealing target for new therapies. Although no targeted therapy has yet been approved for the treatment of KRAS-mutant NSCLC, there are multiple potential therapeutic approaches. These may include direct inhibition of KRAS protein, inhibition of KRAS regulators, alteration of KRAS membrane localization, and inhibition of effector molecules downstream of mutant KRAS. This article provides an overview of the KRAS pathway in lung cancer and related therapeutic strategies under investigation. IMPLICATIONS FOR PRACTICE The identification of oncogene-addicted cancers and specific inhibitors has revolutionized non-small cell lung cancer (NSCLC) treatment and outcomes. One of the most commonly mutated genes in adenocarcinoma is KRAS, found in approximately 30% of lung adenocarcinomas, and thus it is an appealing target for new therapies. This review provides an overview of the KRAS pathway and related targeted therapies under investigation in NSCLC. Some of these agents may play a key role in KRAS-mutant NSCLC treatment in the future.
Collapse
Affiliation(s)
- Pascale Tomasini
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Preet Walia
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Catherine Labbe
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Kevin Jao
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Natasha B Leighl
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Sun J, Chen Y, Li K, Huang Y, Fu X, Zhang X, Zhao W, Wei Y, Xu L, Zhang P, Venkataramanan R, Li S. A prodrug micellar carrier assembled from polymers with pendant farnesyl thiosalicylic acid moieties for improved delivery of paclitaxel. Acta Biomater 2016; 43:282-291. [PMID: 27422196 PMCID: PMC5012958 DOI: 10.1016/j.actbio.2016.07.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/28/2016] [Accepted: 07/11/2016] [Indexed: 11/25/2022]
Abstract
UNLABELLED In order to achieve enhanced and synergistic delivery of paclitaxel (PTX), a hydrophobic anticancer agent, two novel prodrug copolymers, POEG15-b-PFTS6 and POEG15-b-PFTS16 composed of hydrophilic poly(oligo(ethylene glycol) methacrylate) (POEG) and hydrophobic farnesylthiosalicylate (FTS, a nontoxic Ras antagonist) blocks, were synthesized. Both POEG-b-PFTS polymers were able to form micelles with intrinsic antitumor activity in vitro and in vivo. Employing these micelles as a carrier to load PTX, their drug loading capacity, stability, in vivo biodistribution and tumor inhibition effect were evaluated. PTX/POEG15-b-PFTS16 mixed micelles exhibited an excellent stability of 9days at 4°C with a PTX loading capacity of 8.2%, which was more effective than PTX/POEG15-b-PFTS6 mixed micelles. In vivo biodistribution data showed that DiR-loaded POEG-b-PFTS micelles were more effectively localized in the tumor than in other organs. Moreover, both PTX/POEG-b-PFTS micelles showed significantly higher antitumor activity than Taxol in a 4T1.2 murine breast tumor model, and the tumor inhibition and animal survival followed the order of PTX/POEG15-b-PFTS16>PTX/POEG15-b-PFTS6>POEG15-b-PFTS16>Taxol≈POEG15-b-PFTS6. Our data suggest that POEG-b-PFTS micelles are a promising anticancer drug carrier that warrants more studies in the future. STATEMENT OF SIGNIFICANCE Polymerization of drug-based monomer represents a facile and precise method to obtain well-defined polymeric prodrug amphiphiles. Currently, most reports largely focus on the synthesis methods and the biophysical properties. There is limited information about their anti-tumor activity and delivery function as prodrug carriers in vitro and in vivo. In this manuscript, we report the development of two novel prodrug copolymers, POEG15-b-PFTS6 and POEG15-b-PFTS16 composed of hydrophilic poly(oligo(ethylene glycol) methacrylate) (POEG) and hydrophobic farnesylthiosalicylate (FTS, a nontoxic Ras antagonist) blocks. Both POEG-b-PFTS polymers were able to self-assemble into nano-sized micelles with intrinsic antitumor activity in vitro and in vivo. More importantly, POEG-b-PFTS polymers were effective in forming stable mixed micelles with various anticancer agents including PTX, DOX, docetaxel, gefitinib, and imatinib. Delivery of PTX via our new carrier led to significantly improved antitumor activity, suggesting effective PTX/FTS combination therapy. We believe that our study shall be of broad interest to the readers in the fields of biomaterials and drug delivery.
Collapse
Affiliation(s)
- Jingjing Sun
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yichao Chen
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ke Li
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA; Department of Radiation Oncology, University of Kansas, Lawrence, KS 66045, USA; University of Kansas Cancer Center, University of Kansas, Lawrence, KS 66045, USA
| | - Yixian Huang
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiaofeng Fu
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiaolan Zhang
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Wenchen Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yuan Wei
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Liang Xu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA; Department of Radiation Oncology, University of Kansas, Lawrence, KS 66045, USA; University of Kansas Cancer Center, University of Kansas, Lawrence, KS 66045, USA
| | - Peijun Zhang
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Song Li
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
41
|
The association between let-7, RAS and HIF-1α in Ewing Sarcoma tumor growth. Oncotarget 2016; 6:33834-48. [PMID: 26393682 PMCID: PMC4741806 DOI: 10.18632/oncotarget.5616] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 08/23/2015] [Indexed: 11/25/2022] Open
Abstract
Ewing Sarcoma (ES) is the second most common primary malignant bone tumor in children and adolescents. microRNAs (miRNAs) are involved in cancer as tumor suppressors or oncogenes. We studied the involvement of miRNAs located on chromosomes 11q and 22q that participate in the most common translocation in ES. Of these, we focused on 3 that belong to the let-7 family. We studied the expression levels of let-7a, and let-7b and detected a significant correlation between low expression of let-7b and increased risk of relapse. let-7 is known to be a negative regulator of the RAS oncogene. Indeed, we detected an inverse association between the expression of let-7 and RAS protein levels and its downstream target p-ERK, following transfection of let-7 mimics and inhibitors. Furthermore, we identified let-7 as a negative regulator of HIF-1α and EWS-FLI-1. Moreover, we were able to show that HIF-1α directly binds to the EWS-FLI-1 promoter. Salirasib treatment in-vitro resulted in the reduction of cell viability, migration ability, and in the decrease of cells in S-phase. A significant reduction in tumor burden and in the expression levels of both HIF-1α and EWS-FLI-1 proteins were observed in mice after treatment. Our results support the hypothesis that let-7 is a tumor suppressor that negatively regulates RAS, also in ES, and that HIF-1α may contribute to the aggressive metastatic behavior of ES. Moreover, the reduction in the tumor burden in a mouse model of ES following Salirasib treatment, suggests therapeutic potential for this RAS inhibitor in ES.
Collapse
|
42
|
McCarthy M, Prakash P, Gorfe AA. Computational allosteric ligand binding site identification on Ras proteins. Acta Biochim Biophys Sin (Shanghai) 2016; 48:3-10. [PMID: 26487442 DOI: 10.1093/abbs/gmv100] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/16/2015] [Indexed: 12/19/2022] Open
Abstract
A number of computational techniques have been proposed to expedite the process of allosteric ligand binding site identification in inherently flexible and hence challenging drug targets. Some of these techniques have been instrumental in the discovery of allosteric ligand binding sites on Ras proteins, a group of elusive anticancer drug targets. This review provides an overview of these techniques and their application to Ras proteins. A summary of molecular docking and binding site identification is provided first, followed by a more detailed discussion of two specific techniques for binding site identification in ensembles of Ras conformations generated by molecular simulations.
Collapse
Affiliation(s)
- Michael McCarthy
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Priyanka Prakash
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Alemayehu A Gorfe
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
43
|
Zhang J, Park D, Shin DM, Deng X. Targeting KRAS-mutant non-small cell lung cancer: challenges and opportunities. Acta Biochim Biophys Sin (Shanghai) 2016; 48:11-6. [PMID: 26578706 DOI: 10.1093/abbs/gmv118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022] Open
Abstract
Oncogenic mutations in Kirsten rat sarcoma viral oncogene homolog (KRAS) occur in 15%-30% of non-small cell lung cancer (NSCLC). However, despite decades of intensive research, there is still no direct KRAS inhibitor with clinically proven efficacy. Considering its association with poor treatment response and prognosis of lung cancer, developing an effective inhibitory approach is urgently needed. Here, we review different strategies currently being explored to target KRAS-mutant NSCLC, discuss opportunities and challenges, and also propose some novel methods and concepts with the promise of clinical application.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Dongkyoo Park
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Dong M Shin
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Xingming Deng
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
44
|
Fitzgerald TL, Lertpiriyapong K, Cocco L, Martelli AM, Libra M, Candido S, Montalto G, Cervello M, Steelman L, Abrams SL, McCubrey JA. Roles of EGFR and KRAS and their downstream signaling pathways in pancreatic cancer and pancreatic cancer stem cells. Adv Biol Regul 2015; 59:65-81. [PMID: 26257206 DOI: 10.1016/j.jbior.2015.06.003] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/04/2015] [Indexed: 01/06/2023]
Abstract
Pancreatic cancer is currently the fourth most common cancer, is increasing in incidence and soon will be the second leading cause of cancer death in the USA. This is a deadly malignancy with an incidence that approximates the mortality with 44,000 new cases and 36,000 deaths each year. Surgery, although only modestly successful, is the only curative option. However, due the locally aggressive nature and early metastasis, surgery can be performed on less than 20% of patients. Cytotoxic chemotherapy is palliative, has significant toxicity and improves survival very little. Thus new treatment paradigms are needed desperately. Due to the extremely high frequency of KRAS gene mutations (>90%) detected in pancreatic cancer patients, the roles of the epidermal growth factor receptor (EGFR), Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTORC1/GSK-3 pathways have been investigated in pancreatic cancer for many years. Constitutively active Ras can activate both of these pathways and there is cross talk between Ras and EGFR which is believed to be important in driving metastasis. Mutant KRAS may also drive the expression of GSK-3 through Raf/MEK/ERK-mediated effects on GSK-3 transcription. GSK-3 can then regulate the expression of NF-kappaB which is important in modulating pancreatic cancer chemoresistance. While the receptors and many downstream signaling molecules have been identified and characterized, there is still much to learn about these pathways and how their deregulation can lead to cancer. Multiple inhibitors to EGFR, PI3K, mTOR, GSK-3, Raf, MEK and hedgehog (HH) have been developed and are being evaluated in various cancers. Current research often focuses on the role of these pathways in cancer stem cells (CSC), with the goal to identify sites where therapeutic resistance may develop. Relatively novel fields of investigation such as microRNAs and drugs used for other diseases e.g., diabetes, (metformin) and malaria (chloroquine) have provided new information about therapeutic resistance and CSCs. This review will focus on recent advances in the field and how they affect pancreatic cancer research and treatment.
Collapse
Affiliation(s)
- Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology & Functional Genomics, Section of Pathology & Oncology, Via Androne, Catania, Italy, University of Catania, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology & Functional Genomics, Section of Pathology & Oncology, Via Androne, Catania, Italy, University of Catania, Catania, Italy
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Linda Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA.
| |
Collapse
|
45
|
Wolfson E, Schmukler E, Schokoroy ST, Kloog Y, Pinkas-Kramarski R. Enhancing FTS (Salirasib) efficiency via combinatorial treatment. Biol Cell 2015; 107:130-43. [PMID: 25735913 DOI: 10.1111/boc.201400087] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/26/2015] [Indexed: 11/29/2022]
Abstract
The Ras oncogene transmits signals, which regulate various cellular processes including cell motility, differentiation, growth and death. Since Ras signalling is abnormally activated in more than 30% of human cancers, Ras and its downstream signalling pathways are considered good targets for therapeutic interference. Ras is post-translationally modified by the addition of a farnesyl group, which permits its attachment to the plasma membrane. Exploiting this knowledge, a synthetic Ras inhibitor, S-trans, trans-farnesylthiosalicylic acid (FTS; Salirasib), was developed. FTS resembles the farnesylcysteine group of Ras, and acts as an effective Ras antagonist. In the present review, the effect of FTS in combination with various other drugs, as tested in vitro and in vivo, and its therapeutic potential are discussed. As reviewed, FTS cooperates with diverse therapeutic agents, which significantly improves treatment outcome. Therefore, combinations of FTS with other agents have a potential to serve as anti-cancer or anti-inflammatory therapies.
Collapse
Affiliation(s)
- Eya Wolfson
- Department of Neurobiology, Tel-Aviv University, Ramat-Aviv, Israel
| | | | | | | | | |
Collapse
|
46
|
Hatipoglu G, Hock SW, Weiss R, Fan Z, Sehm T, Ghoochani A, Buchfelder M, Savaskan NE, Eyüpoglu IY. Sunitinib impedes brain tumor progression and reduces tumor-induced neurodegeneration in the microenvironment. Cancer Sci 2015; 106:160-70. [PMID: 25458015 PMCID: PMC4399021 DOI: 10.1111/cas.12580] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/25/2014] [Accepted: 11/26/2014] [Indexed: 12/19/2022] Open
Abstract
Malignant gliomas can be counted to the most devastating tumors in humans. Novel therapies do not achieve significant prolonged survival rates. The cancer cells have an impact on the surrounding vital tissue and form tumor zones, which make up the tumor microenvironment. We investigated the effects of sunitinib, a small molecule multitargeted receptor tyrosine kinase inhibitor, on constituents of the tumor microenvironment such as gliomas, astrocytes, endothelial cells, and neurons. Sunitinib has a known anti-angiogenic effect. We found that sunitinib normalizes the aberrant tumor-derived vasculature and reduces tumor vessel pathologies (i.e. auto-loops). Sunitinib has only minor effects on the normal, physiological, non-proliferating vasculature. We found that neurons and astrocytes are protected by sunitinib against glutamate-induced cell death, whereas sunitinib acts as a toxin towards proliferating endothelial cells and tumor vessels. Moreover, sunitinib is effective in inducing glioma cell death. We determined the underlying pathways by which sunitinib operates as a toxin on gliomas and found vascular endothelial growth factor receptor 2 (VEGFR2, KDR/Flk1) as the main target to execute gliomatoxicity. The apoptosis-inducing effect of sunitinib can be mimicked by inhibition of VEGFR2. Knockdown of VEGFR2 can, in part, foster the resistance of glioma cells to receptor tyrosine kinase inhibitors. Furthermore, sunitinib alleviates tumor-induced neurodegeneration. Hence, we tested whether temozolomide treatment could be potentiated by sunitinib application. Here we show that sunitinib can amplify the effects of temozolomide in glioma cells. Thus, our data indicate that combined treatment with temozolomide does not abrogate the effects of sunitinib. In conclusion, we found that sunitinib acts as a gliomatoxic agent and at the same time carries out neuroprotective effects, reducing tumor-induced neurodegeneration. Thus, this report uncovered sunitinib's actions on the brain tumor microenvironment, revealing novel aspects for adjuvant approaches and new clinical assessment criteria when applied to brain tumor patients.
Collapse
Affiliation(s)
- Gökçe Hatipoglu
- Department of Neurosurgery, Universitätsklinikum Erlangen, Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Makovski V, Jacob-Hirsch J, Gefen-Dor C, Shai B, Ehrlich M, Rechavi G, Kloog Y. Analysis of gene expression array in TSC2-deficient AML cells reveals IRF7 as a pivotal factor in the Rheb/mTOR pathway. Cell Death Dis 2014; 5:e1557. [PMID: 25476905 PMCID: PMC4649829 DOI: 10.1038/cddis.2014.502] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/07/2014] [Accepted: 10/13/2014] [Indexed: 12/14/2022]
Abstract
Mutations in tuberous sclerosis (TSC) genes cause the genetic disorder TSC, as well as other neoplasms, including lymphangioleiomyomatosis (LAM) and angiomyolipomas (AMLs). AMLs are benign renal tumors occur both in sporadic LAM and in TSC. As they carry the same mutations, AML cell lines serve as a model for TSC and LAM. Rheb/mammalian target of rapamycin complex 1 (mTORC1) pathway is chronically activated in TSC-deficient cells, and this activation can be diminished using the appropriate inhibitors. Rapamycin (sirolimus) is a known specific inhibitor of mTORC1, whereas S-trans,trans-farnesylthiosalicylic acid (FTS; salirasib) has been shown to inhibit Rheb. To examine the effect of the Rheb/mTOR inhibition pathway, we used human TSC2-deficient AML cells, derived from a LAM patient. FTS indeed inhibited Rheb in these cells and attenuated their proliferation. After comparative treatments with FTS or rapamycin or by re-expression of TSC2, we carried out a gene array analysis. This yielded a substantial number of commonly altered genes, many of which we identified as downstream targets of the interferon (IFN) regulatory factor 7 (IRF7) transcription factor, a central activator of the IFN type 1 immune response. Furthermore, nuclear localization of IRF7 was impaired by each of the three treatments. Interestingly, the phenomena seen on FTS or rapamycin treatment were selective for TSC2-deficient cells. Moreover, knockdown of IRF7 by siRNA mimicked the decrease in number of the abovementioned genes and also inhibited AML cell proliferation. Altogether, these findings support FTS as a potential treatment for TSC and its related pathologies and IRF7 as a novel target for treatment.
Collapse
Affiliation(s)
- V Makovski
- Department of Neurobiology, The George S Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - J Jacob-Hirsch
- Department of Pediatric Hematology-Oncology, Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - C Gefen-Dor
- Department of Pediatric Hematology-Oncology, Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - B Shai
- Department of Cell Research and Immunology, The George S Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - M Ehrlich
- Department of Cell Research and Immunology, The George S Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - G Rechavi
- 1] Department of Pediatric Hematology-Oncology, Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel [2] Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Y Kloog
- Department of Neurobiology, The George S Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
48
|
Zhang X, Huang Y, Zhao W, Liu H, Marquez R, Lu J, Zhang P, Zhang Y, Li J, Gao X, Venkataramanan R, Xu L, Li S. Targeted delivery of anticancer agents via a dual function nanocarrier with an interfacial drug-interactive motif. Biomacromolecules 2014; 15:4326-35. [PMID: 25325795 PMCID: PMC4229023 DOI: 10.1021/bm501339j] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
![]()
We
have developed a dual-function drug carrier, polyethylene glycol
(PEG)-derivatized farnesylthiosalicylate (FTS). Here we report
that incorporation of a drug-interactive motif (Fmoc) into PEG5k–FTS2 led to further improvement in both
drug loading capacity and formulation stability. Doxorubicin (DOX)
formulated in PEG5k–Fmoc–FTS2 showed
sustained release kinetics slower than those of DOX loaded in PEG5k–FTS2. The maximum tolerated dose of DOX-
or paclitaxel (PTX)-loaded PEG5k–Fmoc–FTS2 was significantly higher than that of the free drug. Pharmacokinetics
and biodistribution studies showed that DOX/PEG5k–Fmoc–FTS2 mixed micelles were able to retain DOX in the bloodstream
for a significant amount of time and efficiently deliver the drug
to tumor sites. More importantly, drug (DOX or PTX)-loaded PEG5k–Fmoc–FTS2 led to superior antitumor
activity over other treatments including drugs formulated in PEG5k–FTS2 in breast cancer and prostate cancer
models. Our improved dual function carrier with a built-in drug-interactive
motif represents a simple and effective system for targeted delivery
of anticancer agents.
Collapse
Affiliation(s)
- Xiaolan Zhang
- Center for Pharmacogenetics; ‡Department of Pharmaceutical Sciences, School of Pharmacy; and §University of Pittsburgh Cancer Institute, University of Pittsburgh , Pittsburgh, Pennsylvania 15261, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Cox AD, Der CJ. Ras history: The saga continues. Small GTPases 2014; 1:2-27. [PMID: 21686117 DOI: 10.4161/sgtp.1.1.12178] [Citation(s) in RCA: 536] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 05/17/2010] [Accepted: 05/24/2010] [Indexed: 12/24/2022] Open
Abstract
Although the roots of Ras sprouted from the rich history of retrovirus research, it was the discovery of mutationally activated RAS genes in human cancer in 1982 that stimulated an intensive research effort to understand Ras protein structure, biochemistry and biology. While the ultimate goal has been developing anti-Ras drugs for cancer treatment, discoveries from Ras have laid the foundation for three broad areas of science. First, they focused studies on the origins of cancer to the molecular level, with the subsequent discovery of genes mutated in cancer that now number in the thousands. Second, elucidation of the biochemical mechanisms by which Ras facilitates signal transduction established many of our fundamental concepts of how a normal cell orchestrates responses to extracellular cues. Third, Ras proteins are also founding members of a large superfamily of small GTPases that regulate all key cellular processes and established the versatile role of small GTP-binding proteins in biology. We highlight some of the key findings of the last 28 years.
Collapse
Affiliation(s)
- Adrienne D Cox
- Department of Radiation Oncology; Lineberger Comprehensive Cancer Center; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | | |
Collapse
|
50
|
MENG FANJIE, CAO BIN, FENG ZENGLI, MA SHUNMAO, WANG HAIGANG, LI YANSHU, LI HUI. Knockdown of mutated H-Ras V12 expression induces chemosensitivity of hepatocellular carcinoma cells to cisplatin treatment in vitro and in nude mouse xenografts. Oncol Rep 2014; 32:2023-30. [DOI: 10.3892/or.2014.3466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/26/2014] [Indexed: 11/06/2022] Open
|