1
|
Jakopovic B, Horvatić A, Baranasic J, Car I, Oršolić N, Jakopovich I, Sedić M, Kraljević Pavelić S. Proteomic study of medicinal mushroom extracts reveals antitumor mechanisms in an advanced colon cancer animal model via ribosomal biogenesis, translation, and metabolic pathways. Front Pharmacol 2024; 15:1475102. [PMID: 39494346 PMCID: PMC11528127 DOI: 10.3389/fphar.2024.1475102] [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: 08/02/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction Colorectal cancer ranks as the third most common cancer in both men and women, with approximately 35% of cases being stage IV metastatic at diagnosis. Even with treatment advancements, the survival rates for these patients remain suboptimal. There is a significant focus on developing multi-targeted therapies due to the common issue of drug resistance in standard and targeted cancer treatments. Medicinal mushrooms, both as single compounds and as complex extracts, have undergone extensive research. Numerous types of mushrooms have been shown to be safe, effective inhibitors of cancer pathways and strong enhancers of the immune system. Methods In this study, we performed both qualitative and quantitative proteomic analyses using tandem mass tags (TMT) on CT26 wild type (CT26. WT) colon cancer tissues from Balb/c mice, which were treated with a special blend of medicinal mushroom extracts, either alone or in combination with the chemotherapy drug 5-fluorouracil. Results The results showed a notable increase in survival rates and indicated that medicinal mushroom preparation Agarikon Plus, both alone and combined with 5-fluorouracil or another medicinal mushroom preparation Agarikon.1, impedes multiple key processes in colorectal cancer progression. The analysis of differentially expressed proteins in treated groups was done by use of bioinformatics tools and a decrease in ribosomal biogenesis (e.g., RPS3) and translation processes (e.g., RPL14) as well as an increase in unfolded protein response (e.g., DNAJC3), lipid metabolism (e.g., ACOT7), and the tricarboxylic acid cycle (e.g., FH) were observed. Conclusion The treatment induced various alterations of known biomarkers and protein clusters critical to the progression and prognosis of colorectal cancer, laying a promising foundation for further translational research on this treatment modality.
Collapse
Affiliation(s)
| | - Anita Horvatić
- Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Jurica Baranasic
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Iris Car
- Centre for Applied Bioanthropology, Institute for Anthropological Research, Zagreb, Croatia
| | - Nada Oršolić
- Division of Animal Physiology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | | | - Mirela Sedić
- Centre for Applied Bioanthropology, Institute for Anthropological Research, Zagreb, Croatia
| | | |
Collapse
|
2
|
Kadyr S, Zhuraliyeva A, Yermekova A, Makhambetova A, Kaldybekov DB, Mun EA, Bulanin D, Askarova SN, Umbayev BA. PLGA-PEG Nanoparticles Loaded with Cdc42 Inhibitor for Colorectal Cancer Targeted Therapy. Pharmaceutics 2024; 16:1301. [PMID: 39458630 PMCID: PMC11510643 DOI: 10.3390/pharmaceutics16101301] [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: 08/11/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: An inhibitor of small Rho GTPase Cdc42, CASIN, has been shown to reduce cancer cell proliferation, migration, and invasion, yet it has several limitations, including rapid drug elimination and low bioavailability, which prevents its systemic administration. In this study, we designed and characterized a nanoparticle-based delivery system for CASIN encapsulated within poly(lactide-co-glycolide)-block-poly(ethylene glycol)-carboxylic acid endcap nanoparticles (PLGA-PEG-COOH NPs) for targeted inhibition of Cdc42 activity in colon cancer. Methods: We applied DLS, TEM, and UV-vis spectroscopy methods to characterize the size, polydispersity index, zeta potential, encapsulation efficiency, loading capacity, and in vitro drug release of the synthesized nanoparticles. The CCK-8 cell viability test was used to study colorectal cancer cell growth in vitro. Results: We showed that CASIN-PLGA-PEG-COOH NPs were smooth, spherical, and had a particle size of 86 ± 1 nm, with an encapsulation efficiency of 66 ± 5% and a drug-loading capacity of 5 ± 1%. CASIN was gradually released from NPs, reaching its peak after 24 h, and could effectively inhibit the proliferation of HT-29 (IC50 = 19.55 µM), SW620 (IC50 = 9.33 µM), and HCT116 (IC50 = 10.45 µM) cells in concentrations ranging between 0.025-0.375 mg/mL. CASIN-PLGA-PEG-COOH NPs demonstrated low hemolytic activity with a hemolytic ratio of less than 1% for all tested concentrations. Conclusion: CASIN-PLGA-PEG-COOH NPs have high encapsulation efficiency, sustained drug release, good hemocompatibility, and antitumor activity in vitro. Our results suggest that PLGA-PEG-COOH nanoparticles loaded with CASIN show potential as a targeted treatment for colorectal cancer and could be recommended for further in vivo evaluation.
Collapse
Affiliation(s)
- Sanazar Kadyr
- School of Medicine, Nazarbayev University, 010000 Astana, Kazakhstan; (S.K.); (D.B.)
| | - Altyn Zhuraliyeva
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000 Astana, Kazakhstan; (A.Z.); (A.Y.); (A.M.); (S.N.A.)
| | - Aislu Yermekova
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000 Astana, Kazakhstan; (A.Z.); (A.Y.); (A.M.); (S.N.A.)
| | - Aigerim Makhambetova
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000 Astana, Kazakhstan; (A.Z.); (A.Y.); (A.M.); (S.N.A.)
| | - Daulet B. Kaldybekov
- Department of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, 050040 Almaty, Kazakhstan;
| | - Ellina A. Mun
- School of Sciences and Humanities, Nazarbayev University, 010000 Astana, Kazakhstan;
| | - Denis Bulanin
- School of Medicine, Nazarbayev University, 010000 Astana, Kazakhstan; (S.K.); (D.B.)
| | - Sholpan N. Askarova
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000 Astana, Kazakhstan; (A.Z.); (A.Y.); (A.M.); (S.N.A.)
| | - Bauyrzhan A. Umbayev
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000 Astana, Kazakhstan; (A.Z.); (A.Y.); (A.M.); (S.N.A.)
| |
Collapse
|
3
|
Tang H, Wu H, Jian Y, Ji T, Wu B, Wu Y, Wang P, Cao T. Immune effector dysfunction signatures predict outcomes in patients with colorectal cancer. Int Immunopharmacol 2024; 132:111949. [PMID: 38552290 DOI: 10.1016/j.intimp.2024.111949] [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: 01/06/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Immune effector dysfunction (IED) is mainly manifested as immune exhaustion and senescence, which are the primary obstacles to the success of cancer immunotherapy. In the current study, we characterized the prognostic relevance of IED signatures in patients with colorectal cancer (CRC). METHODS Immunohistochemistry (IHC) data of CRC tissue samples from 41 newly diagnosed patients in our clinical center (HDPH cohort) were used to investigate the prognostic importance of IED signatures. The results were validated by the RNA sequencing data of 372 CRC patients from the Cancer Genome Atlas (TCGA) database. RESULTS In the HDPH cohorts, high Natural Killer (NK) and CD8+ tumor-infiltrating lymphocytes (TILs) were associated with poor overall survival (OS) and relapse-free survival (RFS) in CRC patients. Optimal IED signatures, including high expression of CCR9, ISG20, and low expression of ICOS, and CACNA2D2, predicted poor OS and RFS. Moreover, high-risk scores estimated by a weighted combination of these four IED genes were associated with poor OS and RFS. Notably, risk stratification was constructed by combining risk score and tumor node metastasis (TNM) stage better than TNM stage alone in predicting OS and RFS for CRC patients. The above results were confirmed in the TCGA cohort. CONCLUSION CCR9, ISG20, ICOS, and CACNA2D2 were optimal IED signatures for predicting the outcomes of CRC patients, which might be a potential biomarker for prognostic stratification and designing novel CRC therapy.
Collapse
Affiliation(s)
- Haifeng Tang
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou 516006, China; Department of General Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou 510810, China
| | - Hongsheng Wu
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou 516006, China; Department of General Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou 510810, China
| | - Yueju Jian
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou 516006, China; Department of General Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou 510810, China
| | - Tengfei Ji
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou 516006, China; Department of General Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou 510810, China
| | - Biwen Wu
- Department of Oncology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Yong Wu
- Department of Oncology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China.
| | - Peipei Wang
- Department of Oncology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China; School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China.
| | - Tiansheng Cao
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou 516006, China; Department of General Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou 510810, China.
| |
Collapse
|
4
|
Itaya T, Sano M, Kajiwara I, Oshima Y, Kuramochi T, Kim J, Ichimaru Y, Kitajima O, Masamune A, Ijichi H, Ishii Y, Suzuki T. Mirogabalin improves cancer-associated pain but increases the risk of malignancy in mice with pancreatic cancer. Pain 2023; 164:1545-1554. [PMID: 36701124 DOI: 10.1097/j.pain.0000000000002852] [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: 09/01/2022] [Accepted: 12/08/2022] [Indexed: 01/27/2023]
Abstract
ABSTRACT Mirogabalin, a selective voltage-gated calcium channel α2δ ligand, improves peripheral neuropathic pain; however, its effects on patients with cancers including pancreatic ductal adenocarcinoma (PDAC) remain unknown. We analyzed the effects of mirogabalin on a KPPC ( LSL-KrasG12D/+; Trp53flox/flox; Pdx-1cre/+ ) mouse model of PDAC. Six-week-old KPPC mice received oral mirogabalin (10 mg/kg/day) (n = 10) or vehicle water (n = 14) until the humane end point. Cancer-associated pain was evaluated using the scores of hunching and mouse grimace scale (MGS). Tumor status and plasma cytokine levels were determined using histopathological analysis and cytokine array, respectively. The effects of mirogabalin on the proliferative ability of PDAC cell lines were determined. The scores of the hunching and MGS improved after mirogabalin administration with a decrease in the plasma levels of inflammatory cytokines, such as tumor necrosis factor-α, interleukin-6, and interferon-γ. Although no significant difference in the survival rate was observed, mirogabalin significantly increased pancreatic tumor size and proliferative index of Ki-67 and cyclins. Local arginase-1 + M2-like tumor-associated macrophages and CD31 + tumor blood vessels increased after mirogabalin administration. By contrast, the number of α-smooth muscle actin + cancer-associated fibroblasts, desmoplastic stroma, and CD8 + T cells decreased. Local myeloperoxidase + tumor-associated neutrophils and CD45R + B cells were unaltered. Mirogabalin enhanced the proliferative ability of PDAC cell lines with the upregulation of cyclins and cyclin-dependent kinases; however, it inhibited the potential of pancreatic stellate cells in vitro. Therefore, our results suggest that mirogabalin improves cancer-associated pain but enhances the proliferative potential of PDAC in vitro and in vivo.
Collapse
Affiliation(s)
- Tomoaki Itaya
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Makoto Sano
- Division of Medical Research Planning and Development, Nihon University School of Medicine, Tokyo, Japan
| | - Ichie Kajiwara
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Yukino Oshima
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Tomoya Kuramochi
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Jinsuk Kim
- Division of Medical Research Planning and Development, Nihon University School of Medicine, Tokyo, Japan
| | - Yoshimi Ichimaru
- School of Pharmacy, Shonan University of Medical Sciences, Yokohama, Japan
| | - Osamu Kitajima
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideaki Ijichi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Clinical Nutrition Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukimoto Ishii
- Division of Medical Research Planning and Development, Nihon University School of Medicine, Tokyo, Japan
| | - Takahiro Suzuki
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Soloveva N, Novikova S, Farafonova T, Tikhonova O, Zgoda V. Proteomic Signature of Extracellular Vesicles Associated with Colorectal Cancer. Molecules 2023; 28:molecules28104227. [PMID: 37241967 DOI: 10.3390/molecules28104227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
The proteins of extracellular vesicles (EVs) provide proteomic signatures that reflect molecular features of EV-producing cells, including cancer cells. Detection of cancer cell EV proteins is of great interest due to the development of novel predictive diagnostic approaches. Using targeted mass spectrometry with stable-isotope-labeled peptide standards (SIS), we measured in this study the levels of 34 EV-associated proteins in vesicles and whole lysate derived from the colorectal cancer (CRC) cell lines Caco-2, HT29 and HCT116. We also evaluated the abundance of 13 EV-associated proteins (FN1, TLN1, ITGB3, HSPA8, TUBA4A, CD9, CD63, HSPG2, ITGB1, GNAI2, TSG101, PACSIN2, and CDC42) in EVs isolated from blood plasma samples from 11 CRC patients and 20 healthy volunteers. Downregulation of TLN1, ITGB3, and TUBA4A with simultaneous upregulation of HSPG2 protein were observed in cancer samples compared to healthy controls. The proteomic cargo of the EVs associated with CRC represents a promising source of potential prognostic markers.
Collapse
Affiliation(s)
- Natalia Soloveva
- Orekhovich Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia
| | - Svetlana Novikova
- Orekhovich Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia
| | - Tatiana Farafonova
- Orekhovich Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia
| | - Olga Tikhonova
- Orekhovich Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia
| | - Victor Zgoda
- Orekhovich Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia
| |
Collapse
|
6
|
Guo Z, Hu L, Wang Q, Wang Y, Liu XP, Chen C, Li S, Hu W. Molecular Characterization and Prognosis of Lactate-Related Genes in Lung Adenocarcinoma. Curr Oncol 2023; 30:2845-2861. [PMID: 36975430 PMCID: PMC10047707 DOI: 10.3390/curroncol30030217] [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: 01/27/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 03/02/2023] Open
Abstract
Objective: To explore the lactate-related genes (LRGs) in lung adenocarcinoma (LUAD) by various methods, construct a prognostic model, and explore the relationship between lactate subtypes and the immune tumor microenvironment (TME). Methods: 24 LRGs were collected. The mutation landscape and the prognosis value of LRGs were explored by using The Cancer Genome Atlas (TCGA) data. Consensus clustering analysis was used for different lactate subtype identification. Based on the lactate subtypes, we explore the landscape of TME cell infiltration. A risk-score was calculated by using the LASSO-Cox analysis. A quantitative real-time PCR assay was utilized to validate the expression of characteristic genes in clinical cancer tissues and paracarinoma tissues from LUAD patients. Results: Comparing the normal samples, 18 LRGs were differentially expressed in tumor samples, which revealed that the differential expression of LRGs may be related to Copy Number Variation (CNV) alterations. The two distinct lactate subtypes were defined. Compared to patients in the LRGcluster A group, LUAD patients in the LRGcluster B group achieved better survival. The prognostic model was constructed based on differentially expressed genes (DEGs) via the LASSO-Cox analysis, which showed the accuracy of predicting the prognosis of LUAD patients using the ROC curve. A high-risk score was related to a high immune score, stromal score, and tumor mutation burden (TMB). Patients had better OS with low risk compared with those with high risk. The sensitivities of different risk groups to chemotherapeutic drugs were explored. Finally, the expression of characteristic genes in clinical cancer tissues and paracarinoma tissues from LUAD patients was verified via qRT-PCR. Conclusions: The lactate subtypes were independent prognostic biomarkers in LUAD. Additionally, the difference in the lactate subtypes was an indispensable feature for the individual TME. The comprehensive evaluation of the lactate subtypes in the single tumor would help us to understand the infiltration characteristics of TME and guide immunotherapy strategies.
Collapse
Affiliation(s)
- Zixin Guo
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liwen Hu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan 430071, China
| | - Qingwen Wang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan 430071, China
| | - Yujin Wang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan 430071, China
| | - Xiao-Ping Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Chen Chen
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Human Genetics Resource Preservation Center of Hubei Province, Wuhan 430071, China
| | - Sheng Li
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Human Genetics Resource Preservation Center of Hubei Province, Wuhan 430071, China
- Correspondence: (S.L.); (W.H.)
| | - Weidong Hu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan 430071, China
- Correspondence: (S.L.); (W.H.)
| |
Collapse
|
7
|
Freischel AR, Teer JK, Luddy K, Cunningham J, Artzy-Randrup Y, Epstein T, Tsai KY, Berglund A, Cleveland JL, Gillies RJ, Brown JS, Gatenby RA. Evolutionary Analysis of TCGA Data Using Over- and Under- Mutated Genes Identify Key Molecular Pathways and Cellular Functions in Lung Cancer Subtypes. Cancers (Basel) 2022; 15:18. [PMID: 36612014 PMCID: PMC9817988 DOI: 10.3390/cancers15010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
We identify critical conserved and mutated genes through a theoretical model linking a gene’s fitness contribution to its observed mutational frequency in a clinical cohort. “Passenger” gene mutations do not alter fitness and have mutational frequencies determined by gene size and the mutation rate. Driver mutations, which increase fitness (and proliferation), are observed more frequently than expected. Non-synonymous mutations in essential genes reduce fitness and are eliminated by natural selection resulting in lower prevalence than expected. We apply this “evolutionary triage” principle to TCGA data from EGFR-mutant, KRAS-mutant, and NEK (non-EGFR/KRAS) lung adenocarcinomas. We find frequent overlap of evolutionarily selected non-synonymous gene mutations among the subtypes suggesting enrichment for adaptations to common local tissue selection forces. Overlap of conserved genes in the LUAD subtypes is rare suggesting negative evolutionary selection is strongly dependent on initiating mutational events during carcinogenesis. Highly expressed genes are more likely to be conserved and significant changes in expression (>20% increased/decreased) are common in genes with evolutionarily selected mutations but not in conserved genes. EGFR-mut cancers have fewer average mutations (89) than KRAS-mut (228) and NEK (313). Subtype-specific variation in conserved and mutated genes identify critical molecular components in cell signaling, extracellular matrix remodeling, and membrane transporters. These findings demonstrate subtype-specific patterns of co-adaptations between the defining driver mutation and somatically conserved genes as well as novel insights into epigenetic versus genetic contributions to cancer evolution.
Collapse
Affiliation(s)
- Audrey R. Freischel
- Departments of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jamie K. Teer
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Departments of Tumor Biology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Kimberly Luddy
- Departments of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Institute for Biodiversity and Ecosystem Dynamics, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jessica Cunningham
- Departments of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Yael Artzy-Randrup
- Departments of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Tamir Epstein
- Departments of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Kenneth Y. Tsai
- Departments of Tumor Biology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Departments of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Anders Berglund
- Departments of Tumor Biology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Institute for Biodiversity and Ecosystem Dynamics, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - John L. Cleveland
- Institute for Biodiversity and Ecosystem Dynamics, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Robert J. Gillies
- Departments of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Departments of Pathology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Diagnostic Imaging & Interventional Radiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Joel S. Brown
- Departments of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Robert A. Gatenby
- Departments of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Diagnostic Imaging & Interventional Radiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
8
|
Nasif D, Real S, Roqué M, Branham MT. CDC42 as an epigenetic regulator of ID4 in triple-negative breast tumors. Breast Cancer 2022; 29:562-573. [DOI: 10.1007/s12282-022-01334-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 01/09/2022] [Indexed: 12/01/2022]
|
9
|
CDC42EP3 promotes colorectal cancer through regulating cell proliferation, cell apoptosis and cell migration. Cancer Cell Int 2021; 21:169. [PMID: 33726765 PMCID: PMC7962261 DOI: 10.1186/s12935-021-01845-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background Nowadays, colorectal cancer (CRC) is one of the most commonly diagnosed malignant tumors worldwide, the incidence rate of which is still increasing year by year. Herein, the objective of this study is to investigate whether CDC42EP3 has regulatory effects in CRC. Methods First, CDC42EP3 knockdown cell model based on HCT116 and RKO cell lines was successfully constructed, which was further used for constructing mouse xenotransplantation models. Importantly, effects of CDC42EP3 knockdown on proliferation, colony formation, apoptosis, and migration of CRC were accessed by MTT assay, EdU staining assay, colony formation assay, Flow cytometry, and Transwell assay. Results As the results, we showed that CDC42EP3 was significantly upregulated in CRC, and its high expression was associated with tumor progression. Furthermore, knockdown of CDC42EP3 could inhibit proliferation, colony formation and migration, and promote apoptosis of CRC cells in vitro. In vivo results further confirmed knockdown of CDC42EP3 attenuated tumor growth in CRC. Interestingly, the regulation of CRC by CDC42EP3 involved not only the change of a variety of apoptosis-related proteins, but also the regulation of downstream signaling pathway. Conclusion In conclusion, the role of CDC42EP3 in CRC was clarified and showed its potential as a target of innovative therapeutic approaches for CRC.
Collapse
|
10
|
Pradhan R, Ngo PA, Martínez-Sánchez LDC, Neurath MF, López-Posadas R. Rho GTPases as Key Molecular Players within Intestinal Mucosa and GI Diseases. Cells 2021; 10:cells10010066. [PMID: 33406731 PMCID: PMC7823293 DOI: 10.3390/cells10010066] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Rho proteins operate as key regulators of the cytoskeleton, cell morphology and trafficking. Acting as molecular switches, the function of Rho GTPases is determined by guanosine triphosphate (GTP)/guanosine diphosphate (GDP) exchange and their lipidation via prenylation, allowing their binding to cellular membranes and the interaction with downstream effector proteins in close proximity to the membrane. A plethora of in vitro studies demonstrate the indispensable function of Rho proteins for cytoskeleton dynamics within different cell types. However, only in the last decades we have got access to genetically modified mouse models to decipher the intricate regulation between members of the Rho family within specific cell types in the complex in vivo situation. Translationally, alterations of the expression and/or function of Rho GTPases have been associated with several pathological conditions, such as inflammation and cancer. In the context of the GI tract, the continuous crosstalk between the host and the intestinal microbiota requires a tight regulation of the complex interaction between cellular components within the intestinal tissue. Recent studies demonstrate that Rho GTPases play important roles for the maintenance of tissue homeostasis in the gut. We will summarize the current knowledge on Rho protein function within individual cell types in the intestinal mucosa in vivo, with special focus on intestinal epithelial cells and T cells.
Collapse
|
11
|
Zhang B, Zhang J, Xia L, Luo J, Zhang L, Xu Y, Zhu X, Chen G. Inhibition of CDC42 reduces macrophage recruitment and suppresses lung tumorigenesis in vivo. J Recept Signal Transduct Res 2020; 41:504-510. [PMID: 32998602 DOI: 10.1080/10799893.2020.1828916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Cell division control (CDC) 42 has been involved in the regulation of diverse cancers. Macrophage recruitment plays an important role in the pathogenesis and development of tumor. However, it remains unclear whether CDC42 contributes to macrophage recruitment and lung tumorigenesis in vivo. METHODS Small interference RNA (siRNA) was used to knock down CDC42 in the Lewis lung carcinoma (LLC)1. The invasion capability of CDC42 knockdown LLC1 cells was evaluated. LLC1 cells with CDC42 targeted small hairpin RNA (shRNA) were inoculated into C57BL/6 mice to establish the tumor-bearing animal model Tumor size and metastasis related proteins were measured. In addition, the invasion of macrophages in the tumor site as well as macrophage chemokine were also determined in the model. RESULTS The capacity of invasion and metastasis of LLC1 cells significantly decreased when CDC42 was knocked down. When inoculated with CDC42 knockdown LLC1 cells in vivo, the tumor size and metastasis related proteins levels both decreased. The invasion capacity of macrophages and the associated macrophage chemokine were also significantly down-regulated. CONCLUSION Our data suggest that the inhibition of CDC42 expression in lung cancer cells can significantly prevent the pathogenesis and development of tumor in an allograft tumor model in vivo, which might provide a novel therapeutic target and potential strategy for lung cancer treatment in the future.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Thoracic Surgery, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Jian Zhang
- Department of Thoracic Surgery, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Lilong Xia
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Jing Luo
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Lei Zhang
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Yanhui Xu
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Xinhai Zhu
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Guoping Chen
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
12
|
Eylem CC, Yilmaz M, Derkus B, Nemutlu E, Camci CB, Yilmaz E, Turkoglu MA, Aytac B, Ozyurt N, Emregul E. Untargeted multi-omic analysis of colorectal cancer-specific exosomes reveals joint pathways of colorectal cancer in both clinical samples and cell culture. Cancer Lett 2019; 469:186-194. [PMID: 31669517 DOI: 10.1016/j.canlet.2019.10.038] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 11/24/2022]
Abstract
Exosomes are naturally secreted nano-vesicles consisting of biochemical molecules including RNAs, metabolites, lipids, and proteins, that emerge as diagnostic tools and disease-specific reporters. Here we offer a systematic and integrative approach for the simultaneous analysis of altered molecules namely metabolites, lipids, and proteins. These components tend to augment the discovery of low abundance signature components, and assist in explanation of molecular basis of colorectal cancer (CRC). In order to investigate CRC-derived exosomes, we selected mi-R19a, miR-21, miR-92a, and miR-1246 positive exosomes for downstream experiments. The overall multi-omic changes were investigated comparatively in cell culture and serum samples. Following a systematic multi-omic study, 37 (cell culture) and 31 (serum) metabolites; 130 (cell culture) and 56 (serum) lipids; 9 (cell culture) and 13 (serum) proteins were seen to be differentially expressed (p < 0.05), enabling discrimination between CRC and control. By using these enriched components, we demonstrated that the joint pathways mainly involving fatty acid and amino acid metabolism related pathways changed in CRC significantly. We conclude that this study increases our understanding of molecular basis of CRC, and provides potential exosomal biomarkers for the non-invasive detection, and discrimination of CRC.
Collapse
Affiliation(s)
- Cemil Can Eylem
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, 06230, Ankara, Turkey
| | - Mehmet Yilmaz
- Department of Chemistry, Science Faculty, Ankara University, 06560, Ankara, Turkey
| | - Burak Derkus
- Biomedical Engineering Department, Faculty of Engineering, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey.
| | - Emirhan Nemutlu
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, 06230, Ankara, Turkey
| | - Can Berk Camci
- Department of Chemistry, Science Faculty, Ankara University, 06560, Ankara, Turkey
| | - Erkan Yilmaz
- Biotechnology Institute, Ankara University, 06560, Ankara, Turkey
| | - Mehmet Akif Turkoglu
- Department of General Surgery, Faculty of Medicine, Gazi University, 06560, Ankara, Turkey
| | - Bulent Aytac
- Department of General Surgery, Faculty of Medicine, Gazi University, 06560, Ankara, Turkey
| | - Neslihan Ozyurt
- Medical Oncology, School of Medicine, Ankara University, 06590, Ankara, Turkey
| | - Emel Emregul
- Department of Chemistry, Science Faculty, Ankara University, 06560, Ankara, Turkey
| |
Collapse
|
13
|
A new optimal gene selection approach for cancer classification using enhanced Jaya-based forest optimization algorithm. Neural Comput Appl 2019. [DOI: 10.1007/s00521-019-04355-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|