1
|
Wu Y, Zhao J, Zhao S, Li J, Luo J, Wang Y. PFKFB4 promotes endometrial cancer by regulating glycolysis through SRC‑3 phosphorylation. Oncol Rep 2025; 53:53. [PMID: 40116122 PMCID: PMC11948970 DOI: 10.3892/or.2025.8886] [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: 12/04/2024] [Accepted: 03/06/2025] [Indexed: 03/23/2025] Open
Abstract
The present study aimed to investigate the role of 6‑phosphofructo‑2‑kinase/fructose‑2,6‑biphosphatase 4 (PFKFB4) in endometrial cancer cells and to explore its potential molecular mechanisms. PFKFB4 expression in endometrial cancer tissues was detected by immunohistochemistry. Cell Counting Kit‑8, Transwell assays and flow cytometry were used to detect cell proliferation, invasion and apoptosis in endometrial cancer cells after PFKFB4 knockdown. An enzyme‑linked immunosorbent assay was used to detect the glucose and lactic acid contents. Western blotting was performed to detect the levels of glycolysis‑related enzymes, steroid receptor coactivator‑3 (SRC‑3), and phosphorylated SRC‑3. In vivo experiments were performed to investigate the tumorigenic potential of PFKFB4. PFKFB4 expression was upregulated in endometrial cancer tissues compared with that in normal controls, and its upregulation was positively correlated with the depth of myometrial invasion, lymph node metastasis, surgical pathological stage and vascular invasion. PFKFB4 knockdown significantly inhibited proliferation and invasion, increased apoptosis, and decreased oxygen consumption and lactic acid production in endometrial cancer cells. PFKFB4 knockdown decreased SRC‑3 phosphorylation. After simultaneous PFKFB4 knockdown and SRC‑3 overexpression in cancer cells, oxygen consumption, lactic acid production, and glycolysis‑related protein expression were increased compared with those in control cells. PFKFB4 knockdown inhibited tumor proliferation, apoptosis and the expression of Ki‑67. PFKFB4 may regulate glycolysis in endometrial cancer cells by targeting SRC‑3, thus promoting endometrial cancer progression.
Collapse
Affiliation(s)
- Yaling Wu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Department of Gynecology and Obstetrics, People's Hospital of Shanxi, Taiyuan, Shanxi 030012, P.R. China
| | - Jianzhen Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shuangshuang Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jianfang Li
- Department of Gynecology and Obstetrics, People's Hospital of Shanxi, Taiyuan, Shanxi 030012, P.R. China
| | - Jin Luo
- Department of Pathology, People's Hospital of Shanxi, Taiyuan, Shanxi 030012, P.R. China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
2
|
Sun Y, Zhu J, Zhong H, Zhang Z, Wang F, Nakamura A, Liu Y, Liu J, Yu J, Zeng G, Lin X, Zhou D, Wu C, Wang L, Deng Y, Wu L. Transcriptome-Wide Association Study Identified Novel Blood Tissue Gene Biomarkers for Prostate Cancer Risk. Prostate 2025; 85:567-579. [PMID: 39878408 DOI: 10.1002/pros.24859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/10/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025]
Abstract
OBJECTIVE A number of susceptibility genes in prostate tissue have been identified to be associated with prostate cancer (PCa) risk. However, the reported genes based on assessing prostate tissue could not fully explain PCa genetic susceptibility. It is believed that genes functioning in the immune system may fill in the gap of some missing heritability. METHODS To study potential susceptibility genes acting in such pathways, we performed a transcriptome-wide association study (TWAS) of 79,194 PCa cases and 61,112 control of European ancestry by using three sets of gene expression prediction models of blood tissue. RESULTS A total of 470 genes were associated at false discovery rates-corrected p-value < 0.05, of which 51 were implicated as likely causal genes based on fine-mapping analysis. Compared with previous literature, 133 novel genes were reported for the first time. Of the identified genes, five (CREB3L4, GSTP1, MAPK3, NKX3-1, and PIK3C2B) were enriched in a PCa signaling pathway, and 128 genes were enriched in five PCa categories. Importantly, 13 genes (SCP2, LMNA, ZNF148, H2AFV, TACC1, FLII, SUPT4H1, CD300LF, MYO9B, COX6B1, CTSA, EP300, and TSPO) showed consistent effect directions for the measured levels in circulating immune cells between PCa cases and controls, and 14 genes (SLC39A1, ZBTB7B, TRIM59, NCEH1, N4BP2, TAGAP, TACC1, TRAF1, AIP, SECTM1, C18orf54, ZNF793, YIF1B, and TSPO) showed consistency for levels in blood exosomes between PCa patients and controls. CONCLUSION The identified blood-based candidate susceptibility genes provide further insights into the genetic basis of PCa risk.
Collapse
Affiliation(s)
- Yanfa Sun
- College of Life Science, Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Provincial Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), Longyan University, Longyan, Fujian, P. R. China
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Jingjing Zhu
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Hua Zhong
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Zichen Zhang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fubo Wang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, P. R. China
| | - Akira Nakamura
- Division of Immunology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yanhui Liu
- College of Life Science, Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Provincial Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), Longyan University, Longyan, Fujian, P. R. China
| | - Jiawen Liu
- College of Life Science, Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Provincial Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), Longyan University, Longyan, Fujian, P. R. China
| | - Jie Yu
- College of Life Science, Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Provincial Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), Longyan University, Longyan, Fujian, P. R. China
| | - Guanghua Zeng
- College of Life Science, Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Provincial Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), Longyan University, Longyan, Fujian, P. R. China
| | - Xin Lin
- College of Life Science, Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Provincial Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), Longyan University, Longyan, Fujian, P. R. China
| | - Dan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chong Wu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Liang Wang
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
3
|
Zhao G, Zeng Y, Cheng W, Karkampouna S, Papadopoulou P, Hu B, Zang S, Wezenberg E, Forn-Cuní G, Lopes-Bastos B, Julio MKD, Kros A, Snaar-Jagalska BE. Peptide-Modified Lipid Nanoparticles Boost the Antitumor Efficacy of RNA Therapeutics. ACS NANO 2025; 19:13685-13704. [PMID: 40176316 PMCID: PMC12004924 DOI: 10.1021/acsnano.4c14625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
RNA therapeutics offer a promising approach to cancer treatment by precisely regulating cancer-related genes. While lipid nanoparticles (LNPs) are currently the most advanced nonviral clinically approved vectors for RNA therapeutics, their antitumor efficacy is limited by their unspecific hepatic accumulation after systemic administration. Thus, there is an urgent need to enhance the delivery efficiency of LNPs to target tumor-residing tissues. Here, we conjugated the cluster of differentiation 44 (CD44)-specific targeting peptide A6 (KPSSPPEE) to the cholesterol of LNPs via PEG, named AKPC-LNP, enabling specific tumor delivery. This modification significantly improved delivery to breast cancer cells both in vitro and in vivo, as shown by flow cytometry and confocal microscopy. We further used AKPC-siYT to codeliver siRNAs targeting the transcriptional coactivators YAP and TAZ, achieving potent gene silencing and increased cell death in both 2D cultures and 3D tumor spheroids, outperforming unmodified LNPs. In a breast tumor cell xenografted zebrafish model, systemically administered AKPC-siYT induced robust silencing of YAP/TAZ and downstream genes and significantly enhanced tumor suppression compared to unmodified LNPs. Additionally, AKPC-siYT effectively reduced proliferation in prostate cancer organoids and tumor growth in a patient-derived xenograft (PDX) model. Overall, we developed highly efficient AKPC-LNPs carrying RNA therapeutics for targeted cancer therapy.
Collapse
Affiliation(s)
- Gangyin Zhao
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
- Shenzhen
Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 51800, China
| | - Ye Zeng
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Wanli Cheng
- Urology
Research Laboratory, Department for BioMedical Research, University of Bern, Bern 3010, Switzerland
| | - Sofia Karkampouna
- Urology
Research Laboratory, Department for BioMedical Research, University of Bern, Bern 3010, Switzerland
- Department
of Urology, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Panagiota Papadopoulou
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Bochuan Hu
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Shuya Zang
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Emma Wezenberg
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Gabriel Forn-Cuní
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Bruno Lopes-Bastos
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Marianna Kruithof-de Julio
- Department
of Urology, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Alexander Kros
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - B. Ewa Snaar-Jagalska
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| |
Collapse
|
4
|
Gama JM, Oliveira RC. CD44 and Its Role in Solid Cancers - A Review: From Tumor Progression to Prognosis and Targeted Therapy. FRONT BIOSCI-LANDMRK 2025; 30:24821. [PMID: 40152366 DOI: 10.31083/fbl24821] [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: 05/13/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 03/29/2025]
Abstract
Cluster of differentiation 44 (CD44) is a transmembrane protein expressed in normal cells but overexpressed in several types of cancer. CD44 plays a major role in tumor progression, both locally and systemically, by direct interaction with the extracellular matrix, inducing tissue remodeling, activation of different cellular pathways, such as Akt or mechanistic target of rapamycin (mTOR), and stimulation of angiogenesis. As a prognostic marker, CD44 has been identified as a major player in cancer stem cells (CSCs). CSCs with a CD44 phenotype are associated with chemoresistance, alone or in combination with other CSC markers, such as CD24 or aldehyde dehydrogenase 1 (ALDH1), and may be used for patient stratification. In the therapy setting, CD44 has been explored as a viable target, directly or indirectly. It has revealed promising potential, paving the way for its future use in the clinical setting. Immunohistochemistry effectively detects CD44 overexpression, enabling patients to be accurately selected for surgery and targeted anti-CD44 therapies. In this review, we highlight the properties of CD44, its expression in normal and tumoral tissues through immunohistochemistry and potential treatment options. We also discuss the clinical significance of this marker and its added value in therapeutic decision-making.
Collapse
Affiliation(s)
- João Martins Gama
- Serviço de Anatomia Patológica, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-313 Porto, Portugal
| | - Rui Caetano Oliveira
- Centro de Investigação em Meio Ambiente, Genética e Oncobiologia-CIMAGO, Faculdade de Medicina, Universidade de Coimbra, 3004-535 Coimbra, Portugal
- Centro de Anatomia Patológica Germano de Sousa, 3000-377 Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
5
|
Drápela S, Kvokačková B, Slabáková E, Kotrbová A, Gömöryová K, Fedr R, Kurfürstová D, Eliáš M, Študent V, Lenčéšová F, Ranjani GS, Pospíchalová V, Bryja V, van Weerden WM, Puhr M, Culig Z, Bouchal J, Souček K. Pre-existing cell subpopulations in primary prostate cancer tumors display surface fingerprints of docetaxel-resistant cells. Cell Oncol (Dordr) 2025; 48:205-218. [PMID: 39162992 PMCID: PMC11850551 DOI: 10.1007/s13402-024-00982-2] [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] [Accepted: 08/07/2024] [Indexed: 08/21/2024] Open
Abstract
PURPOSE Docetaxel resistance is a significant obstacle in the treatment of prostate cancer (PCa), resulting in unfavorable patient prognoses. Intratumoral heterogeneity, often associated with epithelial-to-mesenchymal transition (EMT), has previously emerged as a phenomenon that facilitates adaptation to various stimuli, thus promoting cancer cell diversity and eventually resistance to chemotherapy, including docetaxel. Hence, understanding intratumoral heterogeneity is essential for better patient prognosis and the development of personalized treatment strategies. METHODS To address this, we employed a high-throughput single-cell flow cytometry approach to identify a specific surface fingerprint associated with docetaxel-resistance in PCa cells and complemented it with proteomic analysis of extracellular vesicles. We further validated selected antigens using docetaxel-resistant patient-derived xenografts in vivo and probed primary PCa specimens to interrogate of their surface fingerprint. RESULTS Our approaches revealed a 6-molecule surface fingerprint linked to docetaxel resistance in primary PCa specimens. We observed consistent overexpression of CD95 (FAS/APO-1), and SSEA-4 surface antigens in both in vitro and in vivo docetaxel-resistant models, which was also observed in a cell subpopulation of primary PCa tumors exhibiting EMT features. Furthermore, CD95, along with the essential enzymes involved in SSEA-4 synthesis, ST3GAL1, and ST3GAL2, displayed a significant increase in patients with PCa undergoing docetaxel-based therapy, correlating with poor survival outcomes. CONCLUSION In summary, we demonstrate that the identified 6-molecule surface fingerprint associated with docetaxel resistance pre-exists in a subpopulation of primary PCa tumors before docetaxel treatment. Thus, this fingerprint warrants further validation as a promising predictive tool for docetaxel resistance in PCa patients prior to therapy initiation.
Collapse
Affiliation(s)
- Stanislav Drápela
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno, 612 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital in Brno, Brno, 602 00, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, FL, 33612, Tampa, USA
| | - Barbora Kvokačková
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno, 612 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital in Brno, Brno, 602 00, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
| | - Eva Slabáková
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno, 612 00, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
| | - Anna Kotrbová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
| | - Kristína Gömöryová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
| | - Radek Fedr
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno, 612 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital in Brno, Brno, 602 00, Czech Republic
| | - Daniela Kurfürstová
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, 779 00, Czech Republic
| | - Martin Eliáš
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, 779 00, Czech Republic
| | - Vladimír Študent
- Department of Urology, University Hospital Olomouc, Olomouc, 779 00, Czech Republic
| | - Frederika Lenčéšová
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, 779 00, Czech Republic
| | - Ganji Sri Ranjani
- Central European Institute of Technology, Masaryk University, 625 00, Brno, Czech Republic
| | - Vendula Pospíchalová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
| | - Wytske M van Weerden
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Wytemaweg 80, Rotterdam, 3015 CN, The Netherlands
| | - Martin Puhr
- Department of Urology, Experimental Urology, Medical University of Innsbruck, Anich Strasse 35, Innsbruck, A-6020, Austria
| | - Zoran Culig
- International Clinical Research Center, St. Anne's University Hospital in Brno, Brno, 602 00, Czech Republic
- Department of Urology, Experimental Urology, Medical University of Innsbruck, Anich Strasse 35, Innsbruck, A-6020, Austria
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, 779 00, Czech Republic
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno, 612 00, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital in Brno, Brno, 602 00, Czech Republic.
| |
Collapse
|
6
|
Hindes MT, McElligott AM, Best OG, Ward MP, Selemidis S, Miles MA, Nturubika BD, Gregory PA, Anderson PH, Logan JM, Butler LM, Waugh DJ, O'Leary JJ, Hickey SM, Thurgood LA, Brooks DA. Metabolic reprogramming, malignant transformation and metastasis: Lessons from chronic lymphocytic leukaemia and prostate cancer. Cancer Lett 2025; 611:217441. [PMID: 39755364 DOI: 10.1016/j.canlet.2025.217441] [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: 10/17/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Metabolic reprogramming is a hallmark of cancer, crucial for malignant transformation and metastasis. Chronic lymphocytic leukaemia (CLL) and prostate cancer exhibit similar metabolic adaptations, particularly in glucose and lipid metabolism. Understanding this metabolic plasticity is crucial for identifying mechanisms contributing to metastasis. This review considers glucose and lipid metabolism in CLL and prostate cancer, exploring their roles in healthy and malignant states and during disease progression. In CLL, lipid metabolism supports cell survival and migration, with aggressive disease characterised by increased fatty acid oxidation and altered sphingolipids. Richter's transformation and aggressive lymphoma, however, exhibit a metabolic shift towards increased glycolysis. Similarly, prostate cell metabolism is unique, relying on citrate production in the healthy state and undergoing metabolic reprogramming during malignant transformation. Early-stage prostate cancer cells increase lipid synthesis and uptake, and decrease glycolysis, whereas metastatic cells re-adopt glucose metabolism, likely driven by interactions with the tumour microenvironment. Genetic drivers including TP53 and ATM mutations connect metabolic alterations to disease severity in these two malignancies. The bone microenvironment supports the metabolic demands of these malignancies, serving as an initiation niche for CLL and a homing site for prostate cancer metastases. By comparing these malignancies, this review underscores the importance of metabolic plasticity in cancer progression and highlights how CLL and prostate cancer may be models of circulating and solid tumours more broadly. The metabolic phenotypes throughout cancer cell transformation and metastasis, and the microenvironment in which these processes occur, present opportunities for interventions that could disrupt metastatic processes and improve patient outcomes.
Collapse
Affiliation(s)
- Madison T Hindes
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia.
| | - Anthony M McElligott
- Discipline of Haematology, School of Medicine, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College, Dublin, Ireland
| | - Oliver G Best
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Mark P Ward
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Mark A Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Bukuru D Nturubika
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Paul H Anderson
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Jessica M Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lisa M Butler
- South Australian ImmunoGENomics Cancer Institute and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, Australia; Solid Tumour Program, Precision Cancer Medicine theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - David J Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lauren A Thurgood
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Douglas A Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia; Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
7
|
Yu H, Ning N, He F, Xu J, Zhao H, Duan S, Zhao Y. Targeted Delivery of Geraniol via Hyaluronic Acid-Conjugation Enhances Its Anti-Tumor Activity Against Prostate Cancer. Int J Nanomedicine 2024; 19:155-169. [PMID: 38204602 PMCID: PMC10778230 DOI: 10.2147/ijn.s444815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Background Targeted delivery systems have been developed to improve cancer treatment by reducing side effects and enhancing drug efficacy. Geraniol, a natural product, has demonstrated promising anti-cancer effects in various cancer types, including prostate cancer, which is the most commonly diagnosed cancer in men. Hyaluronic acid (HA), a natural carrier targeting CD44-positive prostate cancer cells, can be utilized in a targeted delivery system. Purpose This study investigated the efficacy of a conjugate of HA and geraniol linked via a disulfide bond linker (HA-SS-Geraniol) in prostate cancer. Materials and Methods The cytotoxicity of HA-SS-Geraniol was evaluated on human PC-3 prostate cancer cells. Flow cytometry was used to assess its effects on mitochondrial membrane potential, apoptosis, and cell cycle arrest. Additionally, proteomic analysis was conducted to explore the underlying mechanism of action induced by HA-SS-Geraniol treatment. A subcutaneous xenograft tumor model was established in nude mice to evaluate the toxicity and efficacy of HA-SS-Geraniol in vivo. Results The results demonstrated that HA-SS-Geraniol exhibited potent cytotoxicity against PC-3 prostate cancer cells by inducing mitochondrial membrane potential loss and apoptosis in vitro. The proteomic analysis further supported the hypothesis that HA-SS-Geraniol induces cell death through mitochondria-mediated apoptosis, as evidenced by differential protein expression. The in vivo mouse model confirmed the safety of HA-SS-Geraniol and its ability to inhibit tumor growth. Conclusion HA-SS-Geraniol holds promise as a biologically safe and potentially effective therapeutic agent for prostate cancer treatment. Its targeted delivery system utilizing HA as a carrier shows potential for improving the efficacy of geraniol in cancer therapy.
Collapse
Affiliation(s)
- Han Yu
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, 325060, People’s Republic of China
- Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou-Kean University, Wenzhou, Zhejiang, 325060, People’s Republic of China
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou-Kean University, Wenzhou, Zhejiang, 325060, People’s Republic of China
- Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University, Union, NJ, 07083, USA
| | - Na Ning
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, 325060, People’s Republic of China
- Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou-Kean University, Wenzhou, Zhejiang, 325060, People’s Republic of China
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou-Kean University, Wenzhou, Zhejiang, 325060, People’s Republic of China
| | - Fujin He
- School of Pharmacy, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Jiao Xu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, People’s Republic of China
| | - Han Zhao
- School of Pharmacy, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Shaofeng Duan
- School of Pharmacy, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
- The First Affiliated Hospital of Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Yunqi Zhao
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, 325060, People’s Republic of China
- Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou-Kean University, Wenzhou, Zhejiang, 325060, People’s Republic of China
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou-Kean University, Wenzhou, Zhejiang, 325060, People’s Republic of China
- Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University, Union, NJ, 07083, USA
| |
Collapse
|
8
|
Hu KF, Shu CW, Lee CH, Tseng CJ, Chou YH, Liu PF. Comparative clinical significance and biological roles of PFKFB family members in oral squamous cell carcinoma. Cancer Cell Int 2023; 23:257. [PMID: 37919747 PMCID: PMC10621127 DOI: 10.1186/s12935-023-03110-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Cancer cells promote glycolysis, which supports rapid cell growth and proliferation. Phosphofructokinase-fructose bisphosphatases (PFKFBs), a family of bidirectional glycolytic enzymes, play key roles in the regulation of glycolysis in many types of cancer. However, their roles in oral squamous cell carcinoma (OSCC), the most common type of oral cancer, are still unknown. METHODS We compared the gene expression levels of PFKFB family members and analyzed their clinical significance in oral cancer patients, whose clinical data were obtained the Cancer Genome Atlas database. Moreover, real-time quantitative polymerase chain reaction, western blotting, assays for cell viability, cell cycle, cell migration and viability of cell spheroid were performed in scramble and PFKFB-silenced cells. RESULTS We discovered that PFKFB3 expression in tumor tissues was slightly higher than that in tumor adjacent normal tissues but that PFKFB4 expression was significantly higher in the tumor tissues of oral cancer patients. High PFKFB3 and PFKFB4 expression had different effects on the prognosis of oral cancer patients with different clinicopathological outcomes. Our data showed that PFKFB3 and PFKFB4 play different roles; PFKFB3 is involved in cell viability, G2/M cell cycle progression, invasion, and migration, whereas PFKFB4 is involved in the drug resistance and cancer stemness of OSCC cells. Furthermore, oral cancer patients with co-expressions of PFKFB3/cell cycle or EMT markers and PFKFB4/stemness markers had poor prognosis. CONCLUSIONS PFKFB3 and PFKFB4 play different biological roles in OSCC cells, which implying that they might be potential prognostic biomarkers for OSCC patients with certain clinicopathological outcomes.
Collapse
Affiliation(s)
- Kai-Fang Hu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Dentistry, Division of Periodontics, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Chih-Wen Shu
- Institute of BioPharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
- Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Cheng-Hsin Lee
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ching-Jiunn Tseng
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
| | - Yu-Hsiang Chou
- Department of Dentistry, Division of Periodontics, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan.
| |
Collapse
|
9
|
Ramakrishnan A, Datta I, Panja S, Patel H, Liu Y, Craige MW, Chu C, Jean-Marie G, Oladoja AR, Kim I, Mitrofanova A. Tissue-specific biological aging predicts progression in prostate cancer and acute myeloid leukemia. Front Oncol 2023; 13:1222168. [PMID: 37746266 PMCID: PMC10512286 DOI: 10.3389/fonc.2023.1222168] [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: 05/16/2023] [Accepted: 08/08/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Chronological aging is a well-recognized diagnostic and prognostic factor in multiple cancer types, yet the role of biological aging in manifesting cancer progression has not been fully explored yet. Methods Given the central role of chronological aging in prostate cancer and AML incidence, here we investigate a tissue-specific role of biological aging in prostate cancer and AML progression. We have employed Cox proportional hazards modeling to associate biological aging genes with cancer progression for patients from specific chronological aging groups and for patients with differences in initial cancer aggressiveness. Results Our prostate cancer-specific investigations nominated four biological aging genes (CD44, GADD45B, STAT3, GFAP) significantly associated with time to disease progression in prostate cancer in Taylor et al. patient cohort. Stratified survival analysis on Taylor dataset and validation on an independent TCGA and DKFZ PRAD patient cohorts demonstrated ability of these genes to predict prostate cancer progression, especially for patients with higher Gleason score and for patients younger than 60 years of age. We have further tested the generalizability of our approach and applied it to acute myeloid leukemia (AML). Our analysis nominated three AML-specific biological aging genes (CDC42EP2, CDC42, ALOX15B) significantly associated with time to AML overall survival, especially for patients with favorable cytogenetic risk score and for patients older than 56 years of age. Discussion Comparison of the identified PC and AML markers to genes selected at random and to known markers of progression demonstrated robustness of our results and nominated the identified biological aging genes as valuable markers of prostate cancer and AML progression, opening new avenues for personalized therapeutic management and potential novel treatment investigations.
Collapse
Affiliation(s)
- Anitha Ramakrishnan
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Indrani Datta
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Sukanya Panja
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Harmony Patel
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Health Informatics and Information Management, College of Applied and Natural Sciences, Louisiana Tech University, Ruston, LA, United States
| | - Yingci Liu
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
- New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Michael W. Craige
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Cassandra Chu
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Giselle Jean-Marie
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Rutgers Youth Enjoy Science Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Abdur-Rahman Oladoja
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Rutgers Youth Enjoy Science Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Isaac Kim
- Department of Urology, Yale School of Medicine, New Haven, CT, United States
| | - Antonina Mitrofanova
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
10
|
Kavitha L, Vijayashree Priyadharsini J, Kattula D, Rao UKM, Balaji Srikanth R, Kuzhalmozhi M, Ranganathan K. Expression of CD44 in Head and Neck Squamous Cell Carcinoma-An In-Silico Study. Glob Med Genet 2023; 10:221-228. [PMID: 37593530 PMCID: PMC10431972 DOI: 10.1055/s-0043-1772459] [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] [Indexed: 08/19/2023] Open
Abstract
Introduction CD44, a multistructural and multifunctional transmembrane glycoprotein, is a promising cancer stem cell (CSC) marker that regulates the properties of CSCs, including self-renewal, tumor initiation, and metastasis, and confers resistance to chemotherapy and radiotherapy. The aim of the present study was to evaluate the gene and protein expression of CD44 and explore its prognostic value in head and neck squamous cell carcinoma (HNSCC). Methodology The present observational study employs computational tools for analysis. The Cancer Genome Atlas Head-Neck Squamous Cell Carcinoma dataset (520 primary HNSCC and 44 normal tissues) from the University of Alabama at Birmingham Cancer platform was used to study the association of CD44 mRNA transcript levels with various clinicopathological characteristics of HNSCC including age, gender, tumor grade, tumor stage, human papillomavirus (HPV) status, p53 mutation status, and overall survival. The CD44 protein expression in HNSCC and normal tissues was ascertained using the National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium Head-and-Neck cancer dataset (108 primary HNSCC and 71 normal tissues). Results CD44 mRNA transcript and protein expression levels were significantly higher in HNSCC tissues than in normal tissues, and high CD44 expression was correlated with poor survival. CD44 was upregulated in Stage 1 and Grade 2 HNSCC compared with other stages and grades. Overexpression of CD44 was observed in HPV-negative and TP53-positive mutant status in HNSCC. Conclusion The pleiotropic roles of CD44 in tumorigenesis urge the need to explore its differential expression in HNSCC. The study concludes that CD44 can be a potential diagnostic and prognostic biomarker for HNSCC and offer new molecular targets for CD44-targeted therapy for cancer management.
Collapse
Affiliation(s)
- Loganathan Kavitha
- Department of Oral and Maxillofacial Pathology, Ragas Dental College and Hospital, ECR, Uthandi, Chennai, Tamil Nadu, India; Affiliated to The Tamil Nadu Dr. MGR Medical University, Guindy, Chennai, Tamil Nadu, India
| | - Jayaseelan Vijayashree Priyadharsini
- Clinical Genetics Lab, Centre for Cellular and Molecular Research (The Blue lab), Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences [SIMATS], Saveetha University, Chennai, Tamil Nadu, India
| | - Deepthi Kattula
- Department of Conservative Dentistry and Endodontics, Ragas Dental College and Hospital, ECR, Uthandi, Chennai, Tamil Nadu, India
| | - Umadevi Krishna Mohan Rao
- Department of Oral and Maxillofacial Pathology, Ragas Dental College and Hospital, ECR, Uthandi, Chennai, Tamil Nadu, India; Affiliated to The Tamil Nadu Dr. MGR Medical University, Guindy, Chennai, Tamil Nadu, India
| | - Rajabather Balaji Srikanth
- Department of Oral and Maxillofacial Surgery, Balaji Dental Clinic, Tambaram West, Tambaram, Chennai, Tamil Nadu, India
| | - Manogaran Kuzhalmozhi
- Department of Pathology, Aringnar Anna Memorial Cancer Research Institute, Kanchipuram, Karapettai, Tamil Nadu, India
| | - Kannan Ranganathan
- Department of Oral and Maxillofacial Pathology, Ragas Dental College and Hospital, ECR, Uthandi, Chennai, Tamil Nadu, India; Affiliated to The Tamil Nadu Dr. MGR Medical University, Guindy, Chennai, Tamil Nadu, India
| |
Collapse
|
11
|
Mitra Ghosh T, Mazumder S, Davis J, Yadav J, Akinpelu A, Alnaim A, Kumar H, Waliagha R, Church Bird AE, Rais-Bahrami S, Bird RC, Mistriotis P, Mishra A, Yates CC, Mitra AK, Arnold RD. Metronomic Administration of Topotecan Alone and in Combination with Docetaxel Inhibits Epithelial-mesenchymal Transition in Aggressive Variant Prostate Cancers. CANCER RESEARCH COMMUNICATIONS 2023; 3:1286-1311. [PMID: 37476073 PMCID: PMC10355222 DOI: 10.1158/2767-9764.crc-22-0427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/22/2023]
Abstract
Prostate cancer is the second leading cause of noncutaneous cancer-related deaths in American men. Androgen deprivation therapy (ADT), radical prostatectomy, and radiotherapy remain the primary treatment for patients with early-stage prostate cancer (castration-sensitive prostate cancer). Following ADT, many patients ultimately develop metastatic castration-resistant prostate cancer (mCRPC). Standard chemotherapy options for CRPC are docetaxel (DTX) and cabazitaxel, which increase median survival, although the development of resistance is common. Cancer stem-like cells possess mesenchymal phenotypes [epithelial-to-mesenchymal transition (EMT)] and play crucial roles in tumor initiation and progression of mCRPC. We have shown that low-dose continuous administration of topotecan (METRO-TOPO) inhibits prostate cancer growth by interfering with key cancer pathway genes. This study utilized bulk and single-cell or whole-transcriptome analysis [(RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq)], and we observed greater expression of several EMT markers, including Vimentin, hyaluronan synthase-3, S100 calcium binding protein A6, TGFB1, CD44, CD55, and CD109 in European American and African American aggressive variant prostate cancer (AVPC) subtypes-mCRPC, neuroendocrine variant (NEPC), and taxane-resistant. The taxane-resistant gene FSCN1 was also expressed highly in single-cell subclonal populations in mCRPC. Furthermore, metronomic-topotecan single agent and combinations with DTX downregulated these EMT markers as well as CD44+ and CD44+/CD133+ "stem-like" cell populations. A microfluidic chip-based cell invasion assay revealed that METRO-TOPO treatment as a single agent or in combination with DTX was potentially effective against invasive prostate cancer spread. Our RNA-seq and scRNA-seq analysis were supported by in silico and in vitro studies, suggesting METRO-TOPO combined with DTX may inhibit oncogenic progression by reducing cancer stemness in AVPC through the inhibition of EMT markers and multiple oncogenic factors/pathways. Significance The utilization of metronomic-like dosing regimens of topotecan alone and in combination with DTX resulted in the suppression of makers associated with EMT and stem-like cell populations in AVPC models. The identification of molecular signatures and their potential to serve as novel biomarkers for monitoring treatment efficacy and disease progression response to treatment efficacy and disease progression were achieved using bulk RNA-seq and single-cell-omics methodologies.
Collapse
Affiliation(s)
- Taraswi Mitra Ghosh
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Division of Urology, Department of Surgery, Mass General Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Suman Mazumder
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Center for Pharmacogenomics and Single-Cell Omics (AUPharmGx), Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Joshua Davis
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Jyoti Yadav
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Ayuba Akinpelu
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, Alabama
| | - Ahmed Alnaim
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Harish Kumar
- Department of Biology and Canter for Cancer Research, Tuskegee University, Tuskegee, Alabama
| | - Razan Waliagha
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Allison E. Church Bird
- Flow Cytometry and High-Speed Cell Sorting Laboratory, Auburn University, Auburn, Alabama
| | - Soroush Rais-Bahrami
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Urology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Radiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - R. Curtis Bird
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Panagiotis Mistriotis
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, Alabama
| | - Amarjit Mishra
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Clayton C. Yates
- Department of Biology and Canter for Cancer Research, Tuskegee University, Tuskegee, Alabama
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Amit K. Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Center for Pharmacogenomics and Single-Cell Omics (AUPharmGx), Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - Robert D. Arnold
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| |
Collapse
|
12
|
Damarasingu PV, Das S, Mh S, Bodapati S. Evaluation of CD44 Expression in Prostatic Adenocarcinoma: An Institutional Study. Cureus 2023; 15:e40510. [PMID: 37461792 PMCID: PMC10350293 DOI: 10.7759/cureus.40510] [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] [Accepted: 06/16/2023] [Indexed: 07/20/2023] Open
Abstract
INTRODUCTION Prostate adenocarcinoma is the second-most common cause of cancer. Globally, many cancer-related deaths among men were noted due to prostate adenocarcinoma. CD44 plays a key role in mediating cell-to-cell and cell-to-matrix interaction, which further helps to maintain the integrity of tissue and also inhibits tumor metastasis. MATERIALS AND METHODS Cross-sectional study was done on chips from transurethral resections of the prostate (TURP) and prostatic core biopsy specimens. All specimens with clinically diagnosed and histopathologically confirmed prostatic adenocarcinoma were included in the study. Prostatic intraepithelial neoplasia (PIN), recurrent cases, and patients who had undergone radiotherapy/ chemotherapy prior to biopsy were excluded from the study. The sample size for the current study was 57 with an 8% prevalence value, 95% confidence interval, and 8% absolute error. Immunoreaction to CD44 antibody is membranous and was evaluated by calculating positively stained cell percentage and staining intensity. These two parameters were added to obtain a final score; a score of 0-3 was considered as negative, and a score of 4-6 was regarded as positive. RESULTS A statistically significant difference was only found between Gleason grade (p<0.001), clinical staging (p<0.002), nodal metastasis (p<0.015), and distant metastasis (p<0.020) with CD44 positive expression. The rest of the parameters like PSA (p=0.642) and age (p=0.051) did not correlate with CD44-positive expression. Out of 29 cases with positive CD44 expression, 100% positivity was seen in Gleason's grades 1, 2, and 3. This indicates that CD44 expression showed lesser positivity in poorly differentiated carcinoma. CD44 positivity was seen in 83.3% in the T2 stage. An inverse relationship between tumor staging and CD44 expression was observed with positive CD44 expression in lower tumor staging which implies loss of CD44 expression was associated with greater tumor aggressiveness. Lymph node metastasis cases showed more negative CD44 expression (59.5%) and the same was noted in patients without distant metastasis, that is in 61% of the subjects. Conclusion: Cells tend to lose the ability of CD44 expression as they progress from well-differentiated adenocarcinoma to poorly differentiated adenocarcinoma. CD44 expression suggests that the tumor is in a well-differentiated and gland-forming state as compared to Gleason's grade. Loss of CD44 expression suggests tumor aggressiveness. Thus, the upregulation of CD44 expression can be considered as a potential target for targeted therapy. As many targeted and gene therapies are in clinical trials, large-scale multicentered studies are needed for a better understanding of the clinical course of the disease.
Collapse
Affiliation(s)
| | - Subhashish Das
- Pathology, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, IND
| | - Soumya Mh
- Pathology, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, IND
| | | |
Collapse
|
13
|
Breusa S, Zilio S, Catania G, Bakrin N, Kryza D, Lollo G. Localized chemotherapy approaches and advanced drug delivery strategies: a step forward in the treatment of peritoneal carcinomatosis from ovarian cancer. Front Oncol 2023; 13:1125868. [PMID: 37287910 PMCID: PMC10242058 DOI: 10.3389/fonc.2023.1125868] [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: 12/16/2022] [Accepted: 05/04/2023] [Indexed: 06/09/2023] Open
Abstract
Peritoneal carcinomatosis (PC) is a common outcome of epithelial ovarian carcinoma and is the leading cause of death for these patients. Tumor location, extent, peculiarities of the microenvironment, and the development of drug resistance are the main challenges that need to be addressed to improve therapeutic outcome. The development of new procedures such as HIPEC (Hyperthermic Intraperitoneal Chemotherapy) and PIPAC (Pressurized Intraperitoneal Aerosol Chemotherapy) have enabled locoregional delivery of chemotherapeutics, while the increasingly efficient design and development of advanced drug delivery micro and nanosystems are helping to promote tumor targeting and penetration and to reduce the side effects associated with systemic chemotherapy administration. The possibility of combining drug-loaded carriers with delivery via HIPEC and PIPAC represents a powerful tool to improve treatment efficacy, and this possibility has recently begun to be explored. This review will discuss the latest advances in the treatment of PC derived from ovarian cancer, with a focus on the potential of PIPAC and nanoparticles in terms of their application to develop new therapeutic strategies and future prospects.
Collapse
Affiliation(s)
- Silvia Breusa
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Apoptosis, Cancer and Development Laboratory- Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Institut PLAsCAN, Centre de Recherche en Cancérologie de Lyon, Institut national de santé et de la recherche médicale (INSERM) U1052-Centre National de la Recherche Scientifique - Unité Mixte de Recherche (CNRS UMR)5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Serena Zilio
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Sociétés d'Accélération du Transfert de Technologies (SATT) Ouest Valorisation, Rennes, France
| | - Giuseppina Catania
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
| | - Naoual Bakrin
- Department of Surgical Oncology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France
- Centre pour l'Innovation en Cancérologie de Lyon (CICLY), Claude Bernard University Lyon 1, Lyon, France
| | - David Kryza
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Imthernat Plateform, Hospices Civils de Lyon, Lyon, France
| | - Giovanna Lollo
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
| |
Collapse
|
14
|
Elbagory AM, Hull R, Meyer M, Dlamini Z. Reports of Plant-Derived Nanoparticles for Prostate Cancer Therapy. PLANTS (BASEL, SWITZERLAND) 2023; 12:plants12091870. [PMID: 37176928 PMCID: PMC10181082 DOI: 10.3390/plants12091870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/18/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023]
Abstract
Plants have demonstrated potential in providing various types of phytomedicines with chemopreventive properties that can combat prostate cancer. However, despite their promising in vitro activity, the incorporation of these phytochemicals into the market as anticancer agents has been hindered by their poor bioavailability, mainly due to their inadequate aqueous solubility, chemical instability, and unsatisfactory circulation time. To overcome these drawbacks, it has been suggested that the incorporation of phytochemicals as nanoparticles can offer a solution. The use of plant-based chemicals can also improve the biocompatibility of the formulated nanoparticles by avoiding the use of certain hazardous chemicals in the synthesis, leading to decreased toxicity in vivo. Moreover, in some cases, phytochemicals can act as targeting agents to tumour sites. This review will focus on and summarize the following points: the different types of nanoparticles that contain individual phytochemicals or plant extracts in their design with the aim of improving the bioavailability of the phytochemicals; the therapeutic evaluation of these nanoparticles against prostate cancer both in vitro and in vivo and the reported mode of action and the different types of anticancer experiments used; how the phytochemicals can also improve the targeting effects of these nanoparticles in some instances; and the potential toxicity of these nanoparticles.
Collapse
Affiliation(s)
- Abdulrahman M Elbagory
- Department of Science and Innovation (DSI)/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Cape Town, Private Bag X17, Bellville 7535, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa
| | - Mervin Meyer
- Department of Science and Innovation (DSI)/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Cape Town, Private Bag X17, Bellville 7535, South Africa
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa
| |
Collapse
|
15
|
Tawara M, Suzuki H, Goto N, Tanaka T, Kaneko MK, Kato Y. A Novel Anti-CD44 Variant 9 Monoclonal Antibody C 44Mab-1 Was Developed for Immunohistochemical Analyses against Colorectal Cancers. Curr Issues Mol Biol 2023; 45:3658-3673. [PMID: 37185762 PMCID: PMC10137259 DOI: 10.3390/cimb45040238] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Cluster of differentiation 44 (CD44) is a type I transmembrane glycoprotein and has been shown to be a cell surface marker of cancer stem-like cells in various cancers. In particular, the splicing variants of CD44 (CD44v) are overexpressed in cancers and play critical roles in cancer stemness, invasiveness, and resistance to chemotherapy and radiotherapy. Therefore, the understanding of the function of each CD44v is indispensable for CD44-targeting therapy. CD44v9 contains the variant 9-encoded region, and its expression predicts poor prognosis in patients with various cancers. CD44v9 plays critical roles in the malignant progression of tumors. Therefore, CD44v9 is a promising target for cancer diagnosis and therapy. Here, we developed sensitive and specific monoclonal antibodies (mAbs) against CD44 by immunizing mice with CD44v3-10-overexpressed Chinese hamster ovary-K1 (CHO/CD44v3-10) cells. We first determined their critical epitopes using enzyme-linked immunosorbent assay and characterized their applications as flow cytometry, western blotting, and immunohistochemistry. One of the established clones, C44Mab-1 (IgG1, kappa), reacted with a peptide of the variant 9-encoded region, indicating that C44Mab-1 recognizes CD44v9. C44Mab-1 could recognize CHO/CD44v3-10 cells or colorectal cancer cell lines (COLO201 and COLO205) in flow cytometric analysis. The apparent dissociation constant (KD) of C44Mab-1 for CHO/CD44v3-10, COLO201, and COLO205 was 2.5 × 10-8 M, 3.3 × 10-8 M, and 6.5 × 10-8 M, respectively. Furthermore, C44Mab-1 was able to detect the CD44v3-10 in western blotting and the endogenous CD44v9 in immunohistochemistry using colorectal cancer tissues. These results indicated that C44Mab-1 is useful for detecting CD44v9 not only in flow cytometry or western blotting but also in immunohistochemistry against colorectal cancers.
Collapse
Affiliation(s)
- Mayuki Tawara
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (M.T.); (N.G.); (T.T.); (M.K.K.)
| | - Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (M.T.); (N.G.); (T.T.); (M.K.K.)
| | - Nohara Goto
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (M.T.); (N.G.); (T.T.); (M.K.K.)
| | - Tomohiro Tanaka
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (M.T.); (N.G.); (T.T.); (M.K.K.)
| | - Mika K. Kaneko
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (M.T.); (N.G.); (T.T.); (M.K.K.)
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Yukinari Kato
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (M.T.); (N.G.); (T.T.); (M.K.K.)
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|
16
|
Asif M, Alvi SS, Azaz T, Khan AR, Tiwari B, Hafeez BB, Nasibullah M. Novel Functionalized Spiro [Indoline-3,5'-pyrroline]-2,2'dione Derivatives: Synthesis, Characterization, Drug-Likeness, ADME, and Anticancer Potential. Int J Mol Sci 2023; 24:ijms24087336. [PMID: 37108498 PMCID: PMC10139052 DOI: 10.3390/ijms24087336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
A highly stereo-selective, one-pot, multicomponent method was chosen to synthesize the novel functionalized 1, 3-cycloaddition spirooxindoles (SOXs) (4a-4h). Synthesized SOXs were analyzed for their drug-likeness and ADME parameters and screened for their anticancer activity. Our molecular docking analysis revealed that among all derivatives of SOXs (4a-4h), 4a has a substantial binding affinity (∆G) -6.65, -6.55, -8.73, and -7.27 Kcal/mol with CD-44, EGFR, AKR1D1, and HER-2, respectively. A functional study demonstrated that SOX 4a has a substantial impact on human cancer cell phenotypes exhibiting abnormality in cytoplasmic and nuclear architecture as well as granule formation leading to cell death. SOX 4a treatment robustly induced reactive oxygen species (ROS) generation in cancer cells as observed by enhanced DCFH-DA signals. Overall, our results suggest that SOX (4a) targets CD-44, EGFR, AKR1D1, and HER-2 and induces ROS generation in cancer cells. We conclude that SOX (4a) could be explored as a potential chemotherapeutic molecule against various cancers in appropriate pre-clinical in vitro and in vivo model systems.
Collapse
Affiliation(s)
- Mohd Asif
- Department of Chemistry, Integral University, Lucknow 226026, Uttar Pradesh, India
| | - Sahir Sultan Alvi
- Department of Immunology and Microbiology, South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Tazeen Azaz
- Department of Biological and Synthetic Chemistry, Centre of Biomedical Research, SGPGIMS-Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Abdul Rahman Khan
- Department of Chemistry, Integral University, Lucknow 226026, Uttar Pradesh, India
| | - Bhoopendra Tiwari
- Department of Biological and Synthetic Chemistry, Centre of Biomedical Research, SGPGIMS-Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Bilal Bin Hafeez
- Department of Immunology and Microbiology, South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Malik Nasibullah
- Department of Chemistry, Integral University, Lucknow 226026, Uttar Pradesh, India
| |
Collapse
|
17
|
Archer Goode E, Wang N, Munkley J. Prostate cancer bone metastases biology and clinical management (Review). Oncol Lett 2023; 25:163. [PMID: 36960185 PMCID: PMC10028493 DOI: 10.3892/ol.2023.13749] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/09/2023] [Indexed: 03/25/2023] Open
Abstract
Prostate cancer (PCa) is one of the most prominent causes of cancer-related mortality in the male population. A highly impactful prognostic factor for patients diagnosed with PCa is the presence or absence of bone metastases. The formation of secondary tumours at the bone is the most commonly observed site for the establishment of PCa metastases and is associated with reduced survival of patients in addition to a cohort of life-debilitating symptoms, including mobility issues and chronic pain. Despite the prevalence of this disease presentation and the high medical relevance of bone metastases, the mechanisms underlying the formation of metastases to the bone and the understanding of what drives the osteotropism exhibited by prostate tumours remain to be fully elucidated. This lack of in-depth understanding manifests in limited effective treatment options for patients with advanced metastatic PCa and culminates in the low rate of survival observed for this sub-set of patients. The present review aims to summarise the most recent promising advances in the understanding of how and why prostate tumours metastasise to the bone, with the ultimate aim of highlighting novel treatment and prognostic targets, which may provide the opportunity to improve the diagnosis and treatment of patients with PCa with bone metastases.
Collapse
Affiliation(s)
- Emily Archer Goode
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, International Centre for Life, Newcastle NE1 3BZ, UK
| | - Ning Wang
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, Sheffield S10 2RX, UK
| | - Jennifer Munkley
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, International Centre for Life, Newcastle NE1 3BZ, UK
| |
Collapse
|
18
|
Glycyl-tRNA Synthetase (GARS) Expression Is Associated with Prostate Cancer Progression and Its Inhibition Decreases Migration, and Invasion In Vitro. Int J Mol Sci 2023; 24:ijms24054260. [PMID: 36901698 PMCID: PMC10001614 DOI: 10.3390/ijms24054260] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
Glycyl-tRNA synthetase (GARS) is a potential oncogene associated with poor overall survival in various cancers. However, its role in prostate cancer (PCa) has not been investigated. Protein expression of GARS was investigated in benign, incidental, advanced, and castrate-resistant PCa (CRPC) patient samples. We also investigated the role of GARS in vitro and validated GARS clinical outcomes and its underlying mechanism, utilizing The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA PRAD) database. Our data revealed a significant association between GARS protein expression and Gleason groups. Knockdown of GARS in PC3 cell lines attenuated cell migration and invasion and resulted in early apoptosis signs and cellular arrest in S phase. Bioinformatically, higher GARS expression was observed in TCGA PRAD cohort, and there was significant association with higher Gleason groups, pathological stage, and lymph nodes metastasis. High GARS expression was also significantly correlated with high-risk genomic aberrations such as PTEN, TP53, FXA1, IDH1, SPOP mutations, and ERG, ETV1, and ETV4 gene fusions. Gene Set Enrichment Analysis (GSEA) of GARS through the TCGA PRAD database provided evidence for upregulation of biological processes such as cellular proliferation. Our findings support the oncogenic role of GARS involved in cellular proliferation and poor clinical outcome and provide further evidence for its use as a potential biomarker in PCa.
Collapse
|
19
|
Naseer F, Ahmad T, Kousar K, Kakar S, Gul R, Anjum S, Shareef U. Formulation for the Targeted Delivery of a Vaccine Strain of Oncolytic Measles Virus (OMV) in Hyaluronic Acid Coated Thiolated Chitosan as a Green Nanoformulation for the Treatment of Prostate Cancer: A Viro-Immunotherapeutic Approach. Int J Nanomedicine 2023; 18:185-205. [PMID: 36643861 PMCID: PMC9838128 DOI: 10.2147/ijn.s386560] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/25/2022] [Indexed: 01/11/2023] Open
Abstract
Background Oncolytic viruses are reported as dynamite against cancer treatment nowadays. Methodology In the present work, a live attenuated oral measles vaccine (OMV) strain was used to formulate a polymeric surface-functionalized ligand-based nanoformulation (NF). OMV (half dose: not less than 500 TCID units; 0.25 mL) was encapsulated in thiolated chitosan and outermost coating with hyaluronic acid by ionic gelation method characterizing parameters was performed. Results and Discussion CD44 high expression was confirmed in prostatic adenocarcinoma (PRAD) by GEPIA which extracted data of normal and cancer tissue from GTEx and TCGA. Bioinformatics tools confirmed the viral hemagglutinin capsid protein interaction with human Caspase-I, NLRP3, and TNF-α and viral fusion protein interaction with COX-II and Caspase-I after successful delivery of MV encapsulated in NFs due to high affinity of hyaluronic acid with CD44 on the surface of prostate cancer cells. Particle size = 275.6 mm, PDI = 0.372, and ±11.5 zeta potential were shown by zeta analysis, while the thiolated group in NFs was confirmed by FTIR and Raman analysis. SEM and XRD showed a spherical smooth surface and crystalline nature, respectively, while TEM confirmed virus encapsulation within nanoparticles, which makes it very useful in targeted virus delivery systems. The virus was released from NFs in a sustained but continuous release pattern till 48 h. The encapsulated virus titer was calculated as 2.34×107 TCID50/mL units, which showed syncytia formation on post-day infection 7. Multiplicities of infection 0.1, 0.5, 1, 3, 5, 10, 15, and 20 of HA-coated OMV-loaded NFs as compared to MV vaccine on PC3 was inoculated with IC50 of 5.1 and 3.52, respectively, and growth inhibition was seen after 72 h via MTT assay which showed apoptotic cancer cell death. Conclusion Active targeted, efficacious, and sustained delivery of formulated oncolytic MV is a potent moiety in cancer treatment at lower doses with safe potential for normal prostate cells.
Collapse
Affiliation(s)
- Faiza Naseer
- Industrial Biotechnology, Atta-ur-Rehman School of Applied Biosciences, National University of Science and Technology, Islamabad, Pakistan
- Shifa College of Pharmaceutical Sciences, Shifa Tameer e Millat University, Islamabad, Pakistan
| | - Tahir Ahmad
- Industrial Biotechnology, Atta-ur-Rehman School of Applied Biosciences, National University of Science and Technology, Islamabad, Pakistan
| | - Kousain Kousar
- Industrial Biotechnology, Atta-ur-Rehman School of Applied Biosciences, National University of Science and Technology, Islamabad, Pakistan
| | - Salik Kakar
- Healthcare Biotechnology, Atta-ur-Rehman School of Applied Biosciences, National University of Science and Technology, Islamabad, Pakistan
| | - Rabia Gul
- Shifa College of Pharmaceutical Sciences, Shifa Tameer e Millat University, Islamabad, Pakistan
| | - Sadia Anjum
- Department of Biology, University of Hail, Hail, Saudia Arabia
| | - Usman Shareef
- Shifa College of Pharmaceutical Sciences, Shifa Tameer e Millat University, Islamabad, Pakistan
| |
Collapse
|
20
|
Choi SYC, Ribeiro CF, Wang Y, Loda M, Plymate SR, Uo T. Druggable Metabolic Vulnerabilities Are Exposed and Masked during Progression to Castration Resistant Prostate Cancer. Biomolecules 2022; 12:1590. [PMID: 36358940 PMCID: PMC9687810 DOI: 10.3390/biom12111590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 08/27/2023] Open
Abstract
There is an urgent need for exploring new actionable targets other than androgen receptor to improve outcome from lethal castration-resistant prostate cancer. Tumor metabolism has reemerged as a hallmark of cancer that drives and supports oncogenesis. In this regard, it is important to understand the relationship between distinctive metabolic features, androgen receptor signaling, genetic drivers in prostate cancer, and the tumor microenvironment (symbiotic and competitive metabolic interactions) to identify metabolic vulnerabilities. We explore the links between metabolism and gene regulation, and thus the unique metabolic signatures that define the malignant phenotypes at given stages of prostate tumor progression. We also provide an overview of current metabolism-based pharmacological strategies to be developed or repurposed for metabolism-based therapeutics for castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Stephen Y. C. Choi
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Caroline Fidalgo Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
- New York Genome Center, New York, NY 10013, USA
| | - Stephen R. Plymate
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
- Geriatrics Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Takuma Uo
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
| |
Collapse
|
21
|
Raina K, Kandhari K, Jain AK, Ravichandran K, Maroni P, Agarwal C, Agarwal R. Stage-Specific Effect of Inositol Hexaphosphate on Cancer Stem Cell Pool during Growth and Progression of Prostate Tumorigenesis in TRAMP Model. Cancers (Basel) 2022; 14:4204. [PMID: 36077751 PMCID: PMC9455012 DOI: 10.3390/cancers14174204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/23/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Herein, we assessed the stage-specific efficacy of inositol hexaphosphate (IP6, phytic acid), a bioactive food component, on prostate cancer (PCa) growth and progression in a transgenic mouse model of prostate cancer (TRAMP). Starting at 4, 12, 20, and 30 weeks of age, male TRAMP mice were fed either regular drinking water or 2% IP6 in water for ~8-15 weeks. Pathological assessments at study endpoint indicated that tumor grade is arrested at earlier stages by IP6 treatment; IP6 also prevented progression to more advanced forms of the disease (~55-70% decrease in moderately and poorly differentiated adenocarcinoma incidence was observed in advanced stage TRAMP cohorts). Next, we determined whether the protective effects of IP6 are mediated via its effect on the expansion of the cancer stem cells (CSCs) pool; results indicated that the anti-PCa effects of IP6 are associated with its potential to eradicate the PCa CSC pool in TRAMP prostate tumors. Furthermore, in vitro assays corroborated the above findings as IP6 decreased the % of floating PC-3 prostaspheres (self-renewal of CSCs) by ~90%. Together, these findings suggest the multifaceted chemopreventive-translational potential of IP6 intervention in suppressing the growth and progression of PCa and controlling this malignancy at an early stage.
Collapse
Affiliation(s)
- Komal Raina
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Kushal Kandhari
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anil K. Jain
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kameswaran Ravichandran
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paul Maroni
- Department of Surgery, Division of Urology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
22
|
Sun J, Jin R. PFKFB4 modulated by miR-195-5p can boost the malignant progression of cervical cancer cells. Bioorg Med Chem Lett 2022; 73:128916. [DOI: 10.1016/j.bmcl.2022.128916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 12/24/2022]
|
23
|
Naseer F, Ahmad T, Kousar K, Kakar S, Gul R, Anjum S. Formulation of surface-functionalized hyaluronic acid-coated thiolated chitosan nano-formulation for the delivery of vincristine in prostate cancer: A multifunctional targeted drug delivery approach. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
24
|
Coley AB, Stahly AN, Kasukurthi MV, Barchie AA, Hutcheson SB, Houserova D, Huang Y, Watters BC, King VM, Dean MA, Roberts JT, DeMeis JD, Amin KV, McInnis CH, Godang NL, Wright RM, Haider DF, Piracha NB, Brown CL, Ijaz ZM, Li S, Xi Y, McDonald OG, Huang J, Borchert GM. MicroRNA-like snoRNA-Derived RNAs (sdRNAs) Promote Castration-Resistant Prostate Cancer. Cells 2022; 11:1302. [PMID: 35455981 PMCID: PMC9032336 DOI: 10.3390/cells11081302] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/06/2022] [Accepted: 04/10/2022] [Indexed: 12/13/2022] Open
Abstract
We have identified 38 specifically excised, differentially expressed snoRNA fragments (sdRNAs) in TCGA prostate cancer (PCa) patient samples as compared to normal prostate controls. SnoRNA-derived fragments sdRNA-D19b and -A24 emerged among the most differentially expressed and were selected for further experimentation. We found that the overexpression of either sdRNA significantly increased PC3 (a well-established model of castration-resistant prostate cancer (CRPC)) cell proliferation, and that sdRNA-D19b overexpression also markedly increased the rate of PC3 cell migration. In addition, both sdRNAs provided drug-specific resistances with sdRNA-D19b levels correlating with paclitaxel resistance and sdRNA-24A conferring dasatinib resistance. In silico and in vitro analyses revealed that two established PCa tumor suppressor genes, CD44 and CDK12, represent targets for sdRNA-D19b and sdRNA-A24, respectively. This outlines a biologically coherent mechanism by which sdRNAs downregulate tumor suppressors in AR-PCa to enhance proliferative and metastatic capabilities and to encourage chemotherapeutic resistance. Aggressive proliferation, rampant metastasis, and recalcitrance to chemotherapy are core characteristics of CRPC that synergize to produce a pathology that ranks second in cancer-related deaths for men. This study defines sdRNA-D19b and -A24 as contributors to AR-PCa, potentially providing novel biomarkers and therapeutic targets of use in PCa clinical intervention.
Collapse
Affiliation(s)
- Alexander B. Coley
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
| | - Ashlyn N. Stahly
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Mohan V. Kasukurthi
- School of Computing, University of South Alabama, Mobile, AL 36608, USA; (M.V.K.); (S.L.); (J.H.)
| | - Addison A. Barchie
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- Department of Biology, University of South Alabama, Mobile, AL 36608, USA;
| | - Sam B. Hutcheson
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
| | - Dominika Houserova
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
| | - Yulong Huang
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
| | - Brianna C. Watters
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
| | - Valeria M. King
- Department of Biology, University of South Alabama, Mobile, AL 36608, USA;
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Meghan A. Dean
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- Department of Biology, University of South Alabama, Mobile, AL 36608, USA;
| | - Justin T. Roberts
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jeffrey D. DeMeis
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- Department of Biology, University of South Alabama, Mobile, AL 36608, USA;
| | - Krisha V. Amin
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
| | - Cameron H. McInnis
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- Department of Biology, University of South Alabama, Mobile, AL 36608, USA;
| | - Noel L. Godang
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- Department of Biology, University of South Alabama, Mobile, AL 36608, USA;
| | - Ryan M. Wright
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- Department of Biology, University of South Alabama, Mobile, AL 36608, USA;
| | - David F. Haider
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- Department of Biology, University of South Alabama, Mobile, AL 36608, USA;
| | - Neha B. Piracha
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- Department of Biology, University of South Alabama, Mobile, AL 36608, USA;
| | - Cana L. Brown
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
| | - Zohaib M. Ijaz
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
| | - Shengyu Li
- School of Computing, University of South Alabama, Mobile, AL 36608, USA; (M.V.K.); (S.L.); (J.H.)
| | - Yaguang Xi
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Oliver G. McDonald
- Department of Pathology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33146, USA;
| | - Jingshan Huang
- School of Computing, University of South Alabama, Mobile, AL 36608, USA; (M.V.K.); (S.L.); (J.H.)
| | - Glen M. Borchert
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36608, USA; (A.B.C.); (A.A.B.); (S.B.H.); (D.H.); (Y.H.); (B.C.W.); (M.A.D.); (J.T.R.); (J.D.D.); (K.V.A.); (C.H.M.); (N.L.G.); (R.M.W.); (D.F.H.); (N.B.P.); (C.L.B.); (Z.M.I.)
- School of Computing, University of South Alabama, Mobile, AL 36608, USA; (M.V.K.); (S.L.); (J.H.)
| |
Collapse
|
25
|
Li D, Tang J, Gao R, Lan J, Shen W, Liu Y, Chen Y, Sun H, Yan J, Nie Y, Luo N. PFKFB4 promotes angiogenesis via IL-6/STAT5A/P-STAT5 signaling in breast cancer. J Cancer 2022; 13:212-224. [PMID: 34976184 PMCID: PMC8692682 DOI: 10.7150/jca.66773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/10/2021] [Indexed: 01/16/2023] Open
Abstract
Breast cancer has become the most newly-diagnosed cancer and the 5th leading cause of cancer death worldwide. The 5-year survival rate of breast cancer is about 90%. However, the 5-year survival rate drops to <30% when metastasis to distant sites occurs. The blood vessel formation (i.e., angiogenesis) plays a crucial role during the metastatic process. In this study, we investigated the role of PFKFB4 in angiogenesis of breast cancer. Employing in vitro HUVEC tube formation or in vivo orthotopic mouse model, and gene editing or specific small inhibitors strategy, and utilizing qPCR, western blot, ELISA, or immunofluorescent/immunohistochemistry staining methods, we found the following: 1) PFKFB4 upregulates IL-6 expression via NF-κB signaling in breast cancer cells; 2) PFKFB4-induced lactate secretion contributes to NF-κB activation in breast cancer cells; 3) IL-6 elicits angiogenesis via STAT5A/P-STAT5 in HUVEC; 4) 5-MPN (a specific PFKFB4 inhibitor) suppresses angiogenesis in vitro and in vivo. Our findings suggest a potential strategy whereby 5-MPN may lead to an improved therapeutic outcome for breast cancer patients.
Collapse
Affiliation(s)
- Dan Li
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jiaping Tang
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Ruifang Gao
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
- Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin 300131, China
| | - Jinxin Lan
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Wenzhi Shen
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Yanhua Liu
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Nankai University, Tianjin 300071, China
| | - Yanan Chen
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Nankai University, Tianjin 300071, China
| | - Hongwei Sun
- Experimental Center of Operations, Chinese People's Armed Police Force Command College, Tianjin 300250, China
| | - Jie Yan
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Nankai University, Tianjin 300071, China
| | - Yongwei Nie
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Na Luo
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Nankai University, Tianjin 300071, China
| |
Collapse
|
26
|
Chen CL, Lin CY, Kung HJ. Targeting Mitochondrial OXPHOS and Their Regulatory Signals in Prostate Cancers. Int J Mol Sci 2021; 22:13435. [PMID: 34948229 PMCID: PMC8708687 DOI: 10.3390/ijms222413435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence suggests that tumor development requires not only oncogene/tumor suppressor mutations to drive the growth, survival, and metastasis but also metabolic adaptations to meet the increasing energy demand for rapid cellular expansion and to cope with the often nutritional and oxygen-deprived microenvironment. One well-recognized strategy is to shift the metabolic flow from oxidative phosphorylation (OXPHOS) or respiration in mitochondria to glycolysis or fermentation in cytosol, known as Warburg effects. However, not all cancer cells follow this paradigm. In the development of prostate cancer, OXPHOS actually increases as compared to normal prostate tissue. This is because normal prostate epithelial cells divert citrate in mitochondria for the TCA cycle to the cytosol for secretion into seminal fluid. The sustained level of OXPHOS in primary tumors persists in progression to an advanced stage. As such, targeting OXPHOS and mitochondrial activities in general present therapeutic opportunities. In this review, we summarize the recent findings of the key regulators of the OXPHOS pathway in prostate cancer, ranging from transcriptional regulation, metabolic regulation to genetic regulation. Moreover, we provided a comprehensive update of the current status of OXPHOS inhibitors for prostate cancer therapy. A challenge of developing OXPHOS inhibitors is to selectively target cancer mitochondria and spare normal counterparts, which is also discussed.
Collapse
Affiliation(s)
- Chia-Lin Chen
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (C.-L.C.); (C.-Y.L.)
| | - Ching-Yu Lin
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (C.-L.C.); (C.-Y.L.)
| | - Hsing-Jien Kung
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; (C.-L.C.); (C.-Y.L.)
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County 350, Taiwan
- Comprehensive Cancer Center, Department of Biochemistry and Molecular Medicine, University of California at Davis, Sacramento, CA 95817, USA
| |
Collapse
|
27
|
Hassn Mesrati M, Syafruddin SE, Mohtar MA, Syahir A. CD44: A Multifunctional Mediator of Cancer Progression. Biomolecules 2021; 11:1850. [PMID: 34944493 PMCID: PMC8699317 DOI: 10.3390/biom11121850] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/23/2021] [Accepted: 11/02/2021] [Indexed: 12/15/2022] Open
Abstract
CD44, a non-kinase cell surface transmembrane glycoprotein, has been widely implicated as a cancer stem cell (CSC) marker in several cancers. Cells overexpressing CD44 possess several CSC traits, such as self-renewal and epithelial-mesenchymal transition (EMT) capability, as well as a resistance to chemo- and radiotherapy. The CD44 gene regularly undergoes alternative splicing, resulting in the standard (CD44s) and variant (CD44v) isoforms. The interaction of such isoforms with ligands, particularly hyaluronic acid (HA), osteopontin (OPN) and matrix metalloproteinases (MMPs), drive numerous cancer-associated signalling. However, there are contradictory results regarding whether high or low CD44 expression is associated with worsening clinicopathological features, such as a higher tumour histological grade, advanced tumour stage and poorer survival rates. Nonetheless, high CD44 expression significantly contributes to enhanced tumourigenic mechanisms, such as cell proliferation, metastasis, invasion, migration and stemness; hence, CD44 is an important clinical target. This review summarises current research regarding the different CD44 isoform structures and their roles and functions in supporting tumourigenesis and discusses CD44 expression regulation, CD44-signalling pathways and interactions involved in cancer development. The clinical significance and prognostic value of CD44 and the potential of CD44 as a therapeutic target in cancer are also addressed.
Collapse
Affiliation(s)
- Malak Hassn Mesrati
- Nanobiotechnology Research Group, Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400 UPM, Selangor, Malaysia;
| | - Saiful Effendi Syafruddin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (S.E.S.); (M.A.M.)
| | - M. Aiman Mohtar
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (S.E.S.); (M.A.M.)
| | - Amir Syahir
- Nanobiotechnology Research Group, Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400 UPM, Selangor, Malaysia;
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400 UPM, Selangor, Malaysia
| |
Collapse
|
28
|
Qu Z, Ren Y, Shen H, Wang H, Shi L, Tong D. Combination Therapy of Metastatic Castration-Recurrent Prostate Cancer: Hyaluronic Acid Decorated, Cabazitaxel-Prodrug and Orlistat Co-Loaded Nano-System. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3605-3616. [PMID: 34447241 PMCID: PMC8384126 DOI: 10.2147/dddt.s306684] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/03/2021] [Indexed: 12/29/2022]
Abstract
Purpose Prostate cancer (PCa) is the second leading cause of cancer-related death among men in developed countries. Cabazitaxel (CBZ) is recommended as one of the most active chemotherapy agents for PCa. This study aimed to develop a hyaluronic acid (HA) decorated, cabazitaxel-prodrug (HA-CBZ) and orlistat (ORL) co-loaded nano-system against the prostate cancer in vitro and in vivo. Methods Cabazitaxel-prodrug was firstly synthesized by conjugating HA with CBZ through the formation of ester bonds. HA contained ORL and CBZ prodrug co-loaded lipid-polymer hybrid nanoparticles (ORL/HA-CBZ/LPNs) were constructed and characterized in terms of particle size, zeta potential, drug loading capacity and stability. The antitumor efficiency and systemic toxicity of LPNs were evaluated in vitro and in vivo. Results The resulting ORL/HA-CBZ/LPNs were 150.9 nm in particle size with narrow distribution and high entrapment efficiency. The minimum combination index of 0.57 was found at a drug ratio of 1:2 (ORL:HA-CBZ, w/w) in the drug co-loaded formulations, indicating the strongest synergism effect. ORL/HA-CBZ/LPNs demonstrated an enhanced in vitro and in vivo antitumor effect compared with single drug loaded LPNs and free drug formulations. Conclusion ORL/HA-CBZ/LPNs showed remarkable synergism cytotoxicity and the best tumor inhibition efficiency in mice with negligible systemic toxicity. ORL/HA-CBZ/LPNs can be highly useful for targeted prostate cancer therapy.
Collapse
Affiliation(s)
- Zhen Qu
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Yuning Ren
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Hongyu Shen
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Huihui Wang
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Lijie Shi
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Deyong Tong
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| |
Collapse
|
29
|
Cao J, Shi D, Zhu L, Song L. Circ_RASGEF1B Promotes LPS-Induced Apoptosis and Inflammatory Response by Targeting MicroRNA-146a-5p/Pdk1 Axis in Septic Acute Kidney Injury Cell Model. Nephron Clin Pract 2021; 145:748-759. [PMID: 34438395 DOI: 10.1159/000517475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/21/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We intended to investigate the function of circular RNA RasGEF domain family member 1B (circ_RASGEF1B) in lipopolysaccharide (LPS)-induced septic acute kidney injury (AKI) cell model and its associated mechanism. METHODS TCMK-1 cells were exposed to 10 μg/mL LPS for 24 h to establish a septic AKI cell model. Mice were intraperitoneally injected with 10 mg/kg LPS to establish a septic AKI mice model. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot assay were used to measure RNA and protein expression, respectively. Cell viability and apoptosis were assessed by 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and flow cytometry. Cell inflammatory response was analyzed using enzyme-linked immunosorbent assay. Dual-luciferase reporter assay was conducted to confirm the predicted target relationship between microRNA-146a-5p (miR-146a-5p) and circ_RASGEF1B or pyruvate dehydrogenase kinase 1 (Pdk1). RESULTS The circ_RASGEF1B level was upregulated in LPS-induced TCMK-1 cells and septic AKI mice models. LPS exposure reduced cell viability and promoted cell apoptosis and inflammatory response partly by upregulating circ_RASGEF1B. Circ_RASGEF1B bound to miR-146a-5p and miR-146a-5p interference partly overturned circ_RASGEF1B silencing-mediated effects in LPS-induced TCMK-1 cells. Pdk1 was a target of miR-146a-5p, and Pdk1 accumulation partly counteracted miR-146a-5p-induced influences in TCMK-1 cells upon LPS stimulation. CONCLUSION Circ_RASGEF1B promoted LPS-induced apoptosis and inflammatory response in renal tubular epithelial cells partly by upregulating Pdk1 via acting as miR-146a-5p sponge.
Collapse
Affiliation(s)
- Jianghong Cao
- Department of Intensive Care Unit, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Dongwu Shi
- Department of Intensive Care Unit, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Lili Zhu
- Department of Intensive Care Unit, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Lu Song
- Department of Intensive Care Unit, Shanxi Provincial People's Hospital, Taiyuan, China
| |
Collapse
|
30
|
Lin J, Chen Z, Li Z, Nong D, Li X, Huang G, Hao N, Liang J, Li W. Screening of hub genes and evaluation of the growth regulatory role of CD44 in metastatic prostate cancer. Oncol Rep 2021; 46:196. [PMID: 34296309 PMCID: PMC8317150 DOI: 10.3892/or.2021.8147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the most common cancer type in men worldwide. Currently, the management of metastatic PCa (mPCa) remains a challenge to urologists. The analysis of hub genes and pathways may facilitate the understanding of the molecular mechanism of PCa. In the present study, to identify the hub genes in the mPCa, the three datasets GSE3325, GSE6919 and GSE38241 were downloaded from the platform of the Gene Expression Omnibus and function enrichment analysis of differentially expressed genes (DEGs) was performed. A total of 168 DEGs were obtained and the DEGs were significantly enriched in ‘cell junction’ and ‘cell adhesion’, among others. The Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis demonstrated that DEGs were enriched in three pathways including ‘focal adhesion’, ‘renal cell carcinoma’ and ‘Hippo signaling pathway’. The results of the protein-protein interaction network revealed that the hub genes in mPCa were separately PTEN, Rac GTPase-activating protein 1, protein regulator of cytokinesis 1, PDZ binding kinase, centromere-associated protein E, NUF2 component of NDC80 kinetochore complex, TPX2 microtubule nucleation factor, SOX2, CD44 and ubiquitin-like with PHD and ring finger domains 1. As a hub gene, CD44 was differentially expressed in PCa, as determined by Oncomine analysis. Further experiments in vivo demonstrated that SB-3CT, a selective matrix metalloproteinase inhibitor that has been reported to block CD44 cleavage and inhibit the downstream signaling pathway, suppressed the tumorigenicity of PCa cells by decreasing the expression levels of pyruvate dehydrogenase kinase 1 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4. Moreover, the combination therapy with SB-3CT and docetaxel was more effective in inhibiting PCa compared with monotherapy. In conclusion, the identification of DEGs and the in vivo experimental results helped to elucidate the molecular mechanisms of PCa and provided a potential strategy for the treatment of PCa.
Collapse
Affiliation(s)
- Junhao Lin
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhi Chen
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zuan Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Deyong Nong
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Ximing Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Guihai Huang
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Nan Hao
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jianbo Liang
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Wei Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
31
|
Saidova AA, Potashnikova DM, Tvorogova AV, Paklina OV, Veliev EI, Knyshinsky GV, Setdikova GR, Rotin DL, Maly IV, Hofmann WA, Vorobjev IA. Myosin 1C isoform A is a novel candidate diagnostic marker for prostate cancer. PLoS One 2021; 16:e0251961. [PMID: 34019593 PMCID: PMC8139512 DOI: 10.1371/journal.pone.0251961] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 05/06/2021] [Indexed: 12/26/2022] Open
Abstract
Early diagnosis of prostate cancer is a challenging issue due to the lack of specific markers. Therefore, a sensitive diagnostic marker that is expressed or upregulated exclusively in prostate cancer cells would facilitate diagnostic procedures and ensure a better outcome. We evaluated the expression of myosin 1C isoform A in 5 prostate cell lines, 41 prostate cancer cases, and 11 benign hyperplasias. We analyzed the expression of 12 surface molecules on prostate cancer cells by flow cytometry and analyzed whether high or low myosin 1C isoform A expression could be attributed to a distinct phenotype of prostate cancer cells. Median myosin 1C isoform A expression in prostate cancer samples and cancer cell lines was 2 orders of magnitude higher than in benign prostate hyperplasia. Based on isoform A expression, we could also distinguish clinical stage 2 from clinical stage 3. Among cell lines, PC-3 cells with the highest myosin 1C isoform A level had diminished numbers of CD10/CD13-positive cells and increased numbers of CD29 (integrin β1), CD38, CD54 (ICAM1) positive cells. The surface phenotype of clinical samples was similar to prostate cancer cell lines with high isoform A expression and could be described as CD10-/CD13- with heterogeneous expression of other markers. Both for cell lines and cancer specimens we observed the strong correlation of high myosin 1C isoform A mRNA expression and elevated levels of CD29 and CD54, suggesting a more adhesive phenotype for cells with high isoform A expression. Compared to normal tissue, prostate cancer samples had also reduced numbers of CD24- and CD38-positive cells. Our data suggest that a high level of myosin 1C isoform A is a specific marker both for prostate cancer cells and prostate cancer cell lines. High expression of isoform A is associated with less activated (CD24/CD38 low) and more adhesive (CD29/CD54 high) surface phenotype compared to benign prostate tissue.
Collapse
Affiliation(s)
- Aleena A. Saidova
- School of Biology, Cell Biology and Histology Department, M.V. Lomonosov Moscow State University, Moscow, Russia
- * E-mail:
| | - Daria M. Potashnikova
- School of Biology, Cell Biology and Histology Department, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Anna V. Tvorogova
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Oxana V. Paklina
- Pathoanatomy Department, S.P. Botkin Clinical Hospital, Moscow, Russia
| | | | | | | | - Daniil L. Rotin
- Pathoanatomy Department, S.P. Botkin Clinical Hospital, Moscow, Russia
| | - Ivan V. Maly
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America
| | - Wilma A. Hofmann
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America
| | - Ivan A. Vorobjev
- School of Biology, Cell Biology and Histology Department, M.V. Lomonosov Moscow State University, Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
32
|
Dzobo K, Sinkala M. Cancer Stem Cell Marker CD44 Plays Multiple Key Roles in Human Cancers: Immune Suppression/Evasion, Drug Resistance, Epithelial-Mesenchymal Transition, and Metastasis. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:313-332. [PMID: 33961518 DOI: 10.1089/omi.2021.0025] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
One of the most frequently utilized cancer stem cell markers in human cancers, including colorectal cancer and breast cancer, is CD44. A glycoprotein, CD44, traverses the cell membrane and binds to many ligands, including hyaluronan, resulting in activation of signaling cascades. There are conflicting data, however, on expression of CD44 in relationship to subtypes of cancers. Moreover, the associations of CD44 expression with drug resistance, immune infiltration, epithelial-mesenchymal transition (EMT), metastasis, and clinical prognosis in several cancer types are not clear and call for further studies. We report here an original study on CD44 expression in several human cancers and its relationship with tumorigenesis. We harnessed data from the publicly available databases, including The Cancer Genome Atlas, Gene Expression Profiling Interactive Analysis, Oncomine, Genomics of Drug Sensitivity in Cancer, and the Tumor Immune Estimation Resource. Our analysis reveals that CD44 expression varies across cancer types and is significantly associated with cancer patients' survival, in gastric and pancreatic cancers (p < 0.05). In addition, CD44 expression is closely linked with immune infiltration and immune suppressive features in pancreatic, colon adenocarcinoma, and stomach cancer. High CD44 expression was significantly correlated with the expression of drug resistance, EMT, and metastasis associated genes. Tumors expressing high CD44 have higher mutation burden and afflict older patients compared to tumors expressing low CD44. Cell lines expressing high CD44 are more resistant to anticancer drugs compared to those expressing low CD44. Protein-protein interaction investigations and functional enrichment analysis showed that CD44 interacts with gene products related to cell-substrate adhesion, migration, platelet activation, and cellular response to stress. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that these genes play key roles in biological adhesion, cell component organization, locomotion, G-α-signaling, and the response to stimulus. In summary, these findings lend evidence for the multiple key roles played by CD44 in tumorigenesis and suggest that CD44 is considered further in future studies of cancer pathogenesis and the search for novel molecular targets and personalized medicine biomarkers in clinical oncology.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Musalula Sinkala
- Division of Computational Biology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| |
Collapse
|
33
|
Kotowski K, Rosik J, Machaj F, Supplitt S, Wiczew D, Jabłońska K, Wiechec E, Ghavami S, Dzięgiel P. Role of PFKFB3 and PFKFB4 in Cancer: Genetic Basis, Impact on Disease Development/Progression, and Potential as Therapeutic Targets. Cancers (Basel) 2021; 13:909. [PMID: 33671514 PMCID: PMC7926708 DOI: 10.3390/cancers13040909] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Glycolysis is a crucial metabolic process in rapidly proliferating cells such as cancer cells. Phosphofructokinase-1 (PFK-1) is a key rate-limiting enzyme of glycolysis. Its efficiency is allosterically regulated by numerous substances occurring in the cytoplasm. However, the most potent regulator of PFK-1 is fructose-2,6-bisphosphate (F-2,6-BP), the level of which is strongly associated with 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase activity (PFK-2/FBPase-2, PFKFB). PFK-2/FBPase-2 is a bifunctional enzyme responsible for F-2,6-BP synthesis and degradation. Four isozymes of PFKFB (PFKFB1, PFKFB2, PFKFB3, and PFKFB4) have been identified. Alterations in the levels of all PFK-2/FBPase-2 isozymes have been reported in different diseases. However, most recent studies have focused on an increased expression of PFKFB3 and PFKFB4 in cancer tissues and their role in carcinogenesis. In this review, we summarize our current knowledge on all PFKFB genes and protein structures, and emphasize important differences between the isoenzymes, which likely affect their kinase/phosphatase activities. The main focus is on the latest reports in this field of cancer research, and in particular the impact of PFKFB3 and PFKFB4 on tumor progression, metastasis, angiogenesis, and autophagy. We also present the most recent achievements in the development of new drugs targeting these isozymes. Finally, we discuss potential combination therapies using PFKFB3 inhibitors, which may represent important future cancer treatment options.
Collapse
Affiliation(s)
- Krzysztof Kotowski
- Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.K.); (K.J.)
| | - Jakub Rosik
- Department of Pathology, Pomeranian Medical University, 71-252 Szczecin, Poland; (J.R.); (F.M.)
| | - Filip Machaj
- Department of Pathology, Pomeranian Medical University, 71-252 Szczecin, Poland; (J.R.); (F.M.)
| | - Stanisław Supplitt
- Department of Genetics, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Daniel Wiczew
- Department of Biochemical Engineering, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland;
- Laboratoire de physique et chimie théoriques, Université de Lorraine, F-54000 Nancy, France
| | - Karolina Jabłońska
- Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.K.); (K.J.)
| | - Emilia Wiechec
- Department of Biomedical and Clinical Sciences (BKV), Division of Cell Biology, Linköping University, Region Östergötland, 581 85 Linköping, Sweden;
- Department of Otorhinolaryngology in Linköping, Anesthetics, Operations and Specialty Surgery Center, Region Östergötland, 581 85 Linköping, Sweden
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Research Institute in Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Piotr Dzięgiel
- Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.K.); (K.J.)
- Department of Physiotherapy, Wroclaw University School of Physical Education, 51-612 Wroclaw, Poland
| |
Collapse
|
34
|
Wang F, Wu X, Li Y, Cao X, Zhang C, Gao Y. PFKFB4 as a promising biomarker to predict a poor prognosis in patients with gastric cancer. Oncol Lett 2021; 21:296. [PMID: 33732372 PMCID: PMC7905623 DOI: 10.3892/ol.2021.12557] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 12/14/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most common types of cancer worldwide. Previous studies have reported that phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 (PFKFB4) functions as an oncoprotein in various types of cancer. However, the association between PFKFB4 and GC remains unclear. The present study analyzed the expression levels of PFKFB4 in 148 GC tissue samples, including 46 tumor tissues with matched adjacent normal tissues, using immunohistochemistry, compared the expression levels of PFKFB4 between GC and adjacent normal tissues, and determined the association between PFKFB4 expression levels and patient clinicopathological characteristics. In addition, survival curves were generated using the Kaplan-Meier (KM) plotter database to evaluate the association between PFKFB4 expression and GC prognosis. The results revealed that PFKFB4 expression was upregulated in GC tissues compared with in adjacent normal tissues. PFKFB4 expression was associated with patient age, tumor size, pathological tumor (pT) stage and tumor-node-metastasis (pTNM) stage, and upregulated expression levels of PFKFB4 were observed in tumor tissues from patients <65 years old (compared with that in patients ≥65 years old), as well as patients with a larger tumor size and an advanced stage (pT and pTNM stage) disease. In addition, KM survival analysis demonstrated that patients with low PFKFB4 expression had a significantly improved overall survival (OS), first progression survival and post-progression survival times compared with those with high PFKFB4 expression. Furthermore, PFKFB4 expression was negatively associated with OS time in patients with late pT and pTNM stage disease. In conclusion, the results of the present study indicated that the upregulated PFKFB4 expression in GC tissues may serve as a biomarker for a more advanced disease and a poor prognosis in patients with GC.
Collapse
Affiliation(s)
- Fang Wang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xiaoting Wu
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China.,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yajun Li
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xiangmei Cao
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Cao Zhang
- Department of General Surgery, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yujing Gao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
35
|
Kader A, Brangsch J, Kaufmann JO, Zhao J, Mangarova DB, Moeckel J, Adams LC, Sack I, Taupitz M, Hamm B, Makowski MR. Molecular MR Imaging of Prostate Cancer. Biomedicines 2020; 9:1. [PMID: 33375045 PMCID: PMC7822017 DOI: 10.3390/biomedicines9010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 02/06/2023] Open
Abstract
This review summarizes recent developments regarding molecular imaging markers for magnetic resonance imaging (MRI) of prostate cancer (PCa). Currently, the clinical standard includes MR imaging using unspecific gadolinium-based contrast agents. Specific molecular probes for the diagnosis of PCa could improve the molecular characterization of the tumor in a non-invasive examination. Furthermore, molecular probes could enable targeted therapies to suppress tumor growth or reduce the tumor size.
Collapse
Affiliation(s)
- Avan Kader
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
- Department of Biology, Chemistry and Pharmacy, Institute of Biology, Freie Universität Berlin, Königin-Luise-Str. 1-3, 14195 Berlin, Germany
| | - Julia Brangsch
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
- Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Königsweg 67, Building 21, 14163 Berlin, Germany
| | - Jan O. Kaufmann
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
- Division 1.5 Protein Analysis, Federal Institute for Materials Research and Testing (BAM), Richard-Willstätter-Str. 11, 12489 Berlin, Germany
- Department of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany
| | - Jing Zhao
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
| | - Dilyana B. Mangarova
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, Building 12, 14163 Berlin, Germany
| | - Jana Moeckel
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
| | - Lisa C. Adams
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
| | - Ingolf Sack
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
| | - Matthias Taupitz
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
| | - Bernd Hamm
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
| | - Marcus R. Makowski
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (J.O.K.); (J.Z.); (D.B.M.); (J.M.); (L.C.A.); (I.S.); (M.T.); (B.H.); (M.R.M.)
- School of Biomedical Engineering and Imaging Sciences, King’s College London, St Thomas’ Hospital Westminster Bridge Road, London SE1 7EH, UK
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, Munich (TUM), Ismaninger Str. 22, 81675 München, Germany
| |
Collapse
|
36
|
Metabolic regulation of prostate cancer heterogeneity and plasticity. Semin Cancer Biol 2020; 82:94-119. [PMID: 33290846 DOI: 10.1016/j.semcancer.2020.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/12/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Metabolic reprogramming is one of the main hallmarks of cancer cells. It refers to the metabolic adaptations of tumor cells in response to nutrient deficiency, microenvironmental insults, and anti-cancer therapies. Metabolic transformation during tumor development plays a critical role in the continued tumor growth and progression and is driven by a complex interplay between the tumor mutational landscape, epigenetic modifications, and microenvironmental influences. Understanding the tumor metabolic vulnerabilities might open novel diagnostic and therapeutic approaches with the potential to improve the efficacy of current tumor treatments. Prostate cancer is a highly heterogeneous disease harboring different mutations and tumor cell phenotypes. While the increase of intra-tumor genetic and epigenetic heterogeneity is associated with tumor progression, less is known about metabolic regulation of prostate cancer cell heterogeneity and plasticity. This review summarizes the central metabolic adaptations in prostate tumors, state-of-the-art technologies for metabolic analysis, and the perspectives for metabolic targeting and diagnostic implications.
Collapse
|
37
|
Chen ML, Lai CJ, Lin YN, Huang CM, Lin YH. Multifunctional nanoparticles for targeting the tumor microenvironment to improve synergistic drug combinations and cancer treatment effects. J Mater Chem B 2020; 8:10416-10427. [PMID: 33112350 DOI: 10.1039/d0tb01733g] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Docetaxel-based chemotherapy for prostate cancer is the clinical standard of care. However, nonspecific targeting, multiple drug resistance, and adverse side effects are common obstacles. Various natural compounds, including epigallocatechin-3-gallate (EGCG) in combination with taxane, have the potential to be developed as anticancer therapeutics. Although synergistic hydrophobic-hydrophilic combination drugs have been used with some success, the main drawbacks of this approach are poor bioavailability, unfavorable pharmacokinetics, and low tissue distribution. To improve their synergistic effect and overcome limitations, we encapsulated EGCG and low-dose docetaxel within TPGS-conjugated hyaluronic acid and fucoidan-based nanoparticles. This approach might facilitate simultaneous target-specific markers at the edge and center of the tumor and then might increase intratumoral drug accumulation. Additionally, the successful release of bioactive combination drugs was regulated by the pH-sensitive nanoparticles and internalization into prostate cancer cells through CD44 and P-selectin ligand recognition, and the inhibition of cell growth via induced G2/M phase cell cycle arrest was observed in in vitro study. In in vivo studies, treatment with cancer-targeted combination drug-loaded nanoparticles significantly attenuated tumor growth and increased M30 protein expression without causing organ damage. Overall, the multifunctional nanoparticle system improved the drugs' synergistic effect, indicating great potential in its development as a prostate cancer treatment.
Collapse
Affiliation(s)
- Mei-Lin Chen
- Department of Pharmacy, Chen Hsin General Hospital, Taipei, Taiwan
| | - Chih-Jen Lai
- Faculty of Pharmacy, National Yang-Ming University, Taipei, Taiwan.
| | - Yi-Nan Lin
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chien-Ming Huang
- Department of Pharmacy, Chen Hsin General Hospital, Taipei, Taiwan
| | - Yu-Hsin Lin
- Faculty of Pharmacy, National Yang-Ming University, Taipei, Taiwan. and Institute of Biopharmaceutical Sciences, Department and Institute of Pharmacology, Center for Advanced Pharmaceutics and Drug Delivery Research, National Yang-Ming University, Taipei, Taiwan and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
38
|
Liu H, Marquez RT, Wu X, Li K, Vadlamani S, Li S, Wang Y, Xu L, Wu D. A non-intrusive evaluation method for tumor-targeting characteristics of nanomedicines based on in vivo near-infrared fluorescence imaging. J Mater Chem B 2020; 7:4751-4757. [PMID: 31389969 DOI: 10.1039/c9tb00882a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We developed a novel evaluation method for tumor-targeting characteristics of nanomedicines, average tumor-targeting index (average TTI) and "area under the tumor-targeting index-time curve" (AUTC) were established as the indicators for tumor targeting of nanomedicines based on NIR fluorescence imaging, which helps real-time monitoring of targeting ability and tumor changes in vivo without culling animals.
Collapse
Affiliation(s)
- Hao Liu
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan Province 646000, P. R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wu L, Yang Y, Guo X, Shu XO, Cai Q, Shu X, Li B, Tao R, Wu C, Nikas JB, Sun Y, Zhu J, Roobol MJ, Giles GG, Brenner H, John EM, Clements J, Grindedal EM, Park JY, Stanford JL, Kote-Jarai Z, Haiman CA, Eeles RA, Zheng W, Long J. An integrative multi-omics analysis to identify candidate DNA methylation biomarkers related to prostate cancer risk. Nat Commun 2020; 11:3905. [PMID: 32764609 PMCID: PMC7413371 DOI: 10.1038/s41467-020-17673-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/28/2020] [Indexed: 12/21/2022] Open
Abstract
It remains elusive whether some of the associations identified in genome-wide association studies of prostate cancer (PrCa) may be due to regulatory effects of genetic variants on CpG sites, which may further influence expression of PrCa target genes. To search for CpG sites associated with PrCa risk, here we establish genetic models to predict methylation (N = 1,595) and conduct association analyses with PrCa risk (79,194 cases and 61,112 controls). We identify 759 CpG sites showing an association, including 15 located at novel loci. Among those 759 CpG sites, methylation of 42 is associated with expression of 28 adjacent genes. Among 22 genes, 18 show an association with PrCa risk. Overall, 25 CpG sites show consistent association directions for the methylation-gene expression-PrCa pathway. We identify DNA methylation biomarkers associated with PrCa, and our findings suggest that specific CpG sites may influence PrCa via regulating expression of candidate PrCa target genes.
Collapse
Affiliation(s)
- Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA.
| | - Yaohua Yang
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xingyi Guo
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiang Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bingshan Li
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ran Tao
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chong Wu
- Department of Statistics, Florida State University, Tallahassee, FL, USA
| | - Jason B Nikas
- Research & Development, Genomix Inc, Minneapolis, MN, USA
| | - Yanfa Sun
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
- College of Life Science, Longyan University, Longyan, Fujian, P. R. China
| | - Jingjing Zhu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Monique J Roobol
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Graham G Giles
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, 207 Bouverie St, Melbourne, VIC, 3010, Australia
- Cancer Epidemiology & Intelligence Division, Cancer Council Victoria, 615 St Kilda Rd, Melbourne, VIC, 3004, Australia
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Esther M John
- Department of Medicine (Oncology) and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Judith Clements
- Australian Prostate Cancer Research Centre-QLD, Institute of Health and Biomedical Innovation and School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia
- Translational Research Institute, Brisbane, QLD, Australia
| | | | - Jong Y Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Zsofia Kote-Jarai
- Division of Genetics and Epidemiology, The Institute of Cancer Research, and The Royal Marsden NHS Foundation Trust, London, UK
| | - Christopher A Haiman
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rosalind A Eeles
- Division of Genetics and Epidemiology, The Institute of Cancer Research, and The Royal Marsden NHS Foundation Trust, London, UK
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
40
|
Li X, Chen Z, Li Z, Huang G, Lin J, Wei Q, Liang J, Li W. The metabolic role of PFKFB4 in androgen-independent growth in vitro and PFKFB4 expression in human prostate cancer tissue. BMC Urol 2020; 20:61. [PMID: 32487245 PMCID: PMC7268689 DOI: 10.1186/s12894-020-00635-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/22/2020] [Indexed: 01/13/2023] Open
Abstract
Background It is well known that androgen-deprivation therapy (ADT) can inevitably drive prostate cancer (PCa) cells into a castration-resistant state. According to the “Warburg effect”, the metabolism of aggressive tumor cells increases significantly. The growth of cancer cells depends on glycolysis, which may be a potential target for cancer control. 6-Phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 (PFKFB4) plays key roles in the proliferation and metastasis of PCa cells. However, there is very limited knowledge on the role of PFKFB4 in the conversion to castration resistance. The present study aimed to determine the changes in glucose consumption and PFKFB4 expression in LNCaP cells and androgen-independent LNCaP (LNCaP-AI) cells during the whole process of androgen-independent growth. Additionally, PFKFB4 expression in human PCa tissues was evaluated. Methods We established an androgen-independent LNCaP-AI cell line derived from LNCaP cells to mimic the traits of castration resistance in vitro. LNCaP-AI and LNCaP cells were cultured in the corresponding medium containing the same amount of glucose. At the end of experiments, the medium supernatant and blank medium were collected, and absorbance was measured. LNCaP-AI and LNCaP cells were harvested to detect PFKFB4 expression by Western blotting. Prostate tissue samples including PCa tissue, carcinoma-adjacent tissue and benign prostatic hyperplasia (BPH) tissue specimens were evaluated for PFKFB4 expression using immunohistochemistry. Results In 18 h supernatant samples, the glucose consumption and lactate secretion of LNCaP-AI cells were higher than those of LNCaP cells. The Western blot results indicated that PFKFB4 expression was increased in LNCaP-AI cells compared with LNCaP cells. Immunohistochemistry revealed that the expression of PFKFB4 in PCa tissue specimens was higher than that in BPH and adjacent tissue specimens. However, the differences in PCa tissue before and after ADT were not statistically significant. Conclusion PFKFB4 may be associated with enhanced glycolysis during the androgen-independent growth of PCa cells in vitro. PFKFB4 may be a marker of PCa progression. Our results provide a rationale for further clinical investigation of PCa treatment focused on controlling PFKFB4 expression.
Collapse
Affiliation(s)
- Ximing Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.,Graduate School, Guangxi Medical University, Nanning, China
| | - Zhi Chen
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zuan Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Guihai Huang
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Junhao Lin
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Qiang Wei
- Department of Pathology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jianbo Liang
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Wei Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
| |
Collapse
|
41
|
Wang G, Li S, Xue K, Dong S. PFKFB4 is critical for the survival of acute monocytic leukemia cells. Biochem Biophys Res Commun 2020; 526:978-985. [PMID: 32299611 DOI: 10.1016/j.bbrc.2020.03.174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 03/20/2020] [Accepted: 03/28/2020] [Indexed: 12/31/2022]
Abstract
Acute myeloid leukemia (AML), which is characterized by an overproliferation of blood cells, is divided into several subtypes in adults and children. Of those subtypes, acute monocytic leukemia (M4/M5, AMoL) is reported to be associated with abnormal gene fusions that result in monocytic cell differentiation being blocked. However, few studies have shown a relationship between cellular metabolism and the initiation of AMoL. Here, we use the open-access database TCGA to analyze the expression of enzymes in the metabolic cycle and find that PFKFB4 is highly expressed in AMoL. Subsequently, knocking down PFKFB4 in THP-1 and U937 cells significantly inhibits cell growth and increases the sensitivity of cells to chemical drug-induced apoptosis. In line with the gene-editing alterations, treatment with a PFKFB4 inhibitor exhibits similar effects on THP-1 and U937 proliferation and apoptosis. In addition, we find that PFKFB4 functions as a reliable target of the epigenetic regulator MLL, which is a well-known modulator in AMoL. Mechanistically, MLL promotes PFKFB4 expression at the transcriptional level through the putative E2F6 binding site in the promoter of the pfkfb4 gene. Taken together, our results suggest PFKFB4 serves as a downstream target of MLL and functions as a potent therapeutic target in AMoL.
Collapse
Affiliation(s)
- Gongai Wang
- Department of Hematology, Jining No.1 People's Hospital, No.6, Jiankang Road, Jining City, Shandong Province, 272011, PR China
| | - Shumei Li
- Department of Hematology, Jining No.1 People's Hospital, No.6, Jiankang Road, Jining City, Shandong Province, 272011, PR China
| | - Kewei Xue
- Department of Oncology, Jining No.1 People's Hospital, No.6, Jiankang Road, Jining City, Shandong Province, 272011, PR China
| | - Shasha Dong
- Department of Hematology, Jining No.1 People's Hospital, No.6, Jiankang Road, Jining City, Shandong Province, 272011, PR China.
| |
Collapse
|
42
|
Chen YC, Yu YH. The potential of brown adipogenesis and browning in porcine bone marrow-derived mesenchymal stem cells1. J Anim Sci 2020; 96:3635-3644. [PMID: 29878130 DOI: 10.1093/jas/sky230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/05/2018] [Indexed: 12/19/2022] Open
Abstract
Brown adipocyte lineage commitment and differentiation are under complex regulation. Brown adipocytes are derived from mesenchymal stem cells (MSC). Whether porcine bone marrow-derived MSC (BM-MSC) possess the potential to differentiate into brown adipocytes remains unclear. In the current study, we evaluated the ability of porcine BM-MSC to differentiate into brown adipocytes and browning of differentiated adipocytes. We found that similar to rodent models, bone morphogenetic protein 7 (BMP7) was able to trigger the commitment of BM-MSC to the brown adipocyte lineage by elevating expression of marker genes, nrf-1, tfam, zic1, and pgc-1α (P < 0.05). The expression of brown adipocyte-specific genes, prdm16, dio2, and cidea, was significantly induced (P < 0.05) in BMP7-treated porcine BM-MSC after hormonal induction of adipogenesis. The UCP2 and UCP3 protein levels in BMP7-treated porcine BM-MSC were higher than the control group after hormonal induction of adipogenesis, accompanied by increased mitochondrial DNA copy number and mitochondria-specific gene expression (P < 0.05). Furthermore, acute norepinephrine stimulation potentiated brown adipocyte-specific mRNA expression (P < 0.05) in differentiated adipocytes. Similarly, UCP2 and UCP3 protein levels were increased in differentiated adipocytes upon acute norepinephrine stimulation. In addition, mitochondrial DNA copy number and mitochondria-specific gene expression were also significantly increased (P < 0.05) in differentiated adipocytes after acute norepinephrine exposure. Taken together, these results demonstrate for the first time that porcine BM-MSC are able to commit to the brown adipocyte lineage and differentiate into brown adipocytes. Differentiated adipocytes derived from porcine BM-MSC have the developmental potential to transdifferentiate into brown-like adipocytes upon norepinephrine stimulation.
Collapse
Affiliation(s)
- Ying-Chu Chen
- Department of Biotechnology and Animal Science, National Ilan University, Yilan City, Yilan, Taiwan
| | - Yu-Hsiang Yu
- Department of Biotechnology and Animal Science, National Ilan University, Yilan City, Yilan, Taiwan
| |
Collapse
|
43
|
Xu X, Chen Y, Fu Q, Ni D, Zhang J, Li X, Lu S. The chemical diversity and structure-based discovery of allosteric modulators for the PIF-pocket of protein kinase PDK1. J Enzyme Inhib Med Chem 2019; 34:361-374. [PMID: 30734603 PMCID: PMC6327997 DOI: 10.1080/14756366.2018.1553167] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/18/2018] [Accepted: 11/19/2018] [Indexed: 01/06/2023] Open
Abstract
Phosphoinositide-dependent protein kinase-1 (PDK1) is an important protein in mediating the PI3K-AKT pathway and is thus identified as a promising target. The catalytic activity of PDK1 is tightly regulated by allosteric modulators, which bind to the PDK1 Interacting Fragment (PIF) pocket of the kinase domain that is topographically distinct from the orthosteric, ATP binding site. Allosteric modulators by attaching to the less conserved PIF-pocket have remarkable advantages such as higher selectivity, less side effect, and lower toxicity. Targeting allosteric PIF-pocket of PDK1 has become the focus of recent attention. In this review, we summarise the current advances in the structure-based discovery of PDK1 allosteric modulators. We will first present the three-dimensional structure of PDK1 and illustrate the allosteric regulatory mechanism of PDK1 through the modulation of the PIF-pocket. Then, the recent advances of PDK1 allosteric modulators targeting the PIF-pocket will be recapitulated detailly according to the structural similarity of allosteric modulators.
Collapse
Affiliation(s)
- Xinyuan Xu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yingyi Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Qiang Fu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Duan Ni
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jian Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xiaolong Li
- Department of Orthopedics, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Shaoyong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
44
|
A Shifty Target: Tumor-Initiating Cells and Their Metabolism. Int J Mol Sci 2019; 20:ijms20215370. [PMID: 31661927 PMCID: PMC6862122 DOI: 10.3390/ijms20215370] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/25/2019] [Accepted: 10/26/2019] [Indexed: 12/20/2022] Open
Abstract
Tumor-initiating cells (TICs), or cancer stem cells, constitute highly chemoresistant, asymmetrically dividing, and tumor-initiating populations in cancer and are thought to play a key role in metastatic and chemoresistant disease. Tumor-initiating cells are isolated from cell lines and clinical samples based on features such as sphere formation in stem cell medium and expression of TIC markers, typically a set of outer membrane proteins and certain transcription factors. Although both bulk tumor cells and TICs show an adaptive metabolic plasticity, TIC metabolism is thought to differ and likely in a tumor-specific and growth condition-dependent pattern. In the context of some common solid tumor diseases, we here review reports on how TIC isolation methods and markers associate with metabolic features, with some focus on oxidative metabolism, including fatty acid and lipid metabolism. These have emerged as significant factors in TIC phenotypes, and in tumor biology as a whole. Other sections address mitochondrial biogenesis and dynamics in TICs, and the influence of the tumor microenvironment. Further elucidation of the complex biology of TICs and their metabolism will require advanced methodologies.
Collapse
|
45
|
Deng L, Gu X, Zeng T, Xu F, Dong Z, Liu C, Chao H. Identification and characterization of biomarkers and their functions for docetaxel-resistant prostate cancer cells. Oncol Lett 2019; 18:3236-3248. [PMID: 31452801 PMCID: PMC6676406 DOI: 10.3892/ol.2019.10623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 06/13/2019] [Indexed: 12/27/2022] Open
Abstract
Docetaxel treatment is a standard chemotherapy strategy for castration-resistant prostate cancer (CRPC), and patients with CRPC eventually develop resistance to treatment. However, little is understood regarding the underlying mechanism of resistance. The present study aimed to identify the underlying crucial genes and regulatory networks associated with docetaxel resistance in prostate cancer using bioinformatics analyses. For this purpose, one expression profile dataset (GSE33455), which included two docetaxel-sensitive and two docetaxel-resistant cell lines, was downloaded from the Gene Expression Omnibus database, and analyses of differential gene expression and function enrichment were performed. A protein-protein interaction (PPI) network was constructed, and the associated hub genes were investigated using the Search Tool for the Retrieval of Interacting Genes/Proteins and Cytoscape software. A total of 756 differentially expression genes (DEGs) were identified, including 509 downregulated and 247 upregulated genes. Enrichment analysis revealed that the DEGs were associated with the interferon-γ-mediated signaling pathway, protein binding, bicellular tight junctions and cancer pathways. Two modules were screened from the PPI network, and the corresponding genes were identified to be largely enriched in the interferon-γ-mediated signaling pathway and the negative regulators of the DExD/H-Box helicase 58/interferon induced with helicase C domain 1 signaling pathway, and enriched in cell-cell adhesion and the Rap1 signaling pathway. Among the ten hub genes, epidermal growth factor receptor, spleen tyrosine kinase (SYK), intracellular adhesion molecule 1 (ICAM1), interleukin (IL)6, CXC motif chemokine ligand 8 (CXCL8), cyclin dependent kinase 1 and CD44 molecule (CD44) were significantly differentially expressed in prostate cancer tissues compared with healthy tissues based on The Cancer Genome Atlas data. The Gene Expression Profiling Interactive Analysis database revealed that ICAM1 was positively associated with IL6 and CXCL8, and epidermal growth factor receptor was positively associated with CD44 and SYK. Additionally, ten hub genes, which were identified to be associated with the drug resistance of docetaxel in prostatic carcinoma in the present study, were predominantly associated with tumor progression and metastasis. Reverse transcription-quantitative PCR analysis performed on docetaxel-sensitive and docetaxel-resistant prostate cancer cell lines demonstrated that certain hub genes, including CDK1, 2′-5′-oligoadenylate synthetase 3, CXCL8 and CDH1, were highly expressed in the docetaxel-resistant cell lines, which confirmed the bioinformatics results. In conclusion, the present study identified a number of important genes that are associated with the molecular mechanism of docetaxel resistance by integrated bioinformatical analysis, and these genes and regulatory networks may assist with identifying potential gene therapy targets for CRPC. Further functional analyses are required to validate the current findings.
Collapse
Affiliation(s)
- Leihong Deng
- Medical Department of The Graduate School, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaopeng Gu
- Department of Orthopedics, Zhoushan Guhechuan Hospital, Zhoushan, Zhejang 316000, P.R. China
| | - Tao Zeng
- Department of Urology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fanghua Xu
- Pathology Department, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhifeng Dong
- Medical Department of The Graduate School, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chao Liu
- Medical Department of The Graduate School, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Haichao Chao
- Laboratory of Clinical Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
46
|
Chu PY, Tsai SC, Ko HY, Wu CC, Lin YH. Co-Delivery of Natural Compounds with a Dual-Targeted Nanoparticle Delivery System for Improving Synergistic Therapy in an Orthotopic Tumor Model. ACS APPLIED MATERIALS & INTERFACES 2019; 11:23880-23892. [PMID: 31192580 DOI: 10.1021/acsami.9b06155] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Various natural compounds including epigallocatechin gallate (EGCG) and curcumin (CU) have potential in developing anticancer therapy. However, their clinical use is commonly limited by instability and low tissue distribution. EGCG and CU combined treatment can improve the efficacy with synergistic effects. To improve the synergistic effect and overcome the limitations of low tissue distribution, we applied a dual cancer-targeted nanoparticle system to co-deliver EGCG and CU. Nanoparticles were composed of hyaluronic acid, fucoidan, and poly(ethylene glycol)-gelatin to encapsulate EGCG and CU. Furthermore, a dual targeting system was established with hyaluronic acid and fucoidan, which were used as agents for targeting CD44 on prostate cancer cells and P-selectin in tumor vasculature, respectively. Their effect and efficacy were investigated in prostate cancer cells and a orthotopic prostate tumor model. The EGCG/CU-loaded nanoparticles bound to prostate cancer cells, which were uptaken more into cells, leading to a better anticancer efficiency compared to the EGCG/CU combination solution. In addition, the releases of EGCG and CU were regulated by their pH value that avoided the premature release. In mice, treatment of the cancer-targeted EGCG/CU-loaded nanoparticles significantly attenuated the orthotopic tumor growth without inducing organ injuries. Overall, the dual-targeted nanoparticle system for the co-delivery of EGCG and CU greatly improved its synergistic effect in cancer therapy, indicating its great potential in developing treatments for prostate cancer therapy.
Collapse
|
47
|
Di R, Yang Z, Xu P, Xu Y. Silencing PDK1 limits hypoxia-induced pulmonary arterial hypertension in mice via the Akt/p70S6K signaling pathway. Exp Ther Med 2019; 18:699-704. [PMID: 31281449 DOI: 10.3892/etm.2019.7627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 06/15/2018] [Indexed: 12/16/2022] Open
Abstract
The present study aimed to investigate the effect of phosphoinositide-dependent protein kinase-1 (PDK1) on hypoxia-induced pulmonary arterial hypertension (PAH). A mouse model of hypoxia-induced PAH was generated using normal or PDK1-knockout mice. Histological analysis and hemodynamic evaluations were performed to identify the progression of PAH. The expression and phosphorylation of PDK1/protein kinase B (Akt) signaling pathway associated proteins were detected by western blot analysis. Increased lung vessel thickness, right ventricular (RV) systolic pressure (RVSP), RV hypertrophy index (RVHI) values [the RV weight-to-left ventricular (LV) plus septum (S) weight ratio] and PDK1 expression were observed in the hypoxia-induced PAH model compared with the normal control. The phosphorylation of AktT308, proline-rich Akt1 substrate 1 (PRAS40) and S6KT229 was also notably increased in the PAH model compared with the control. The changes of proteins were not observed in the hypoxia treated PDK1flox/+ : Tie2-Cre mice. Similarly, the RVSP and RVHI values, and PDK1 expression were reduced in the hypoxia treated PDK1flox/+: Tie2-Cre mice to a level comparable with those in the control, suggesting that PDK1 partial knockout significantly limited hypoxia-induced PAH. The results of the present study indicate that PDK1 is essential for hypoxia-induced PAH through the PDK1/Akt/S6K signaling cascades.
Collapse
Affiliation(s)
- Ruomin Di
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Zhongzhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, Jiangsu 210061, P.R. China
| | - Peng Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Yingjia Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
48
|
Yi M, Ban Y, Tan Y, Xiong W, Li G, Xiang B. 6-Phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 and 4: A pair of valves for fine-tuning of glucose metabolism in human cancer. Mol Metab 2018; 20:1-13. [PMID: 30553771 PMCID: PMC6358545 DOI: 10.1016/j.molmet.2018.11.013] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/12/2022] Open
Abstract
Background Cancer cells favor the use of less efficient glycolysis rather than mitochondrial oxidative phosphorylation to metabolize glucose, even in oxygen-rich conditions, a distinct metabolic alteration named the Warburg effect or aerobic glycolysis. In adult cells, bifunctional 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase (PFKFB) family members are responsible for controlling the steady-state cytoplasmic levels of fructose-2,6-bisphosphate, which allosterically activates 6-phosphofructo-1-kinase, the key enzyme catalyzing the rate-limiting reaction of glycolysis. PFKFB3 and PFKFB4 are the two main isoenzymes overexpressed in various human cancers. Scope of review In this review, we summarize recent findings on the glycolytic and extraglycolytic roles of PFKFB3 and PFKFB4 in cancer progression and discuss potential therapies for targeting of PFKFB3 and PFKFB4. Major conclusions PFKFB3 has the highest kinase activity to shunt glucose toward glycolysis, whereas PFKFB4 has more FBPase-2 activity, redirecting glucose toward the pentose phosphate pathway, providing reducing power for lipid biosynthesis and scavenging reactive oxygen species. Co-expression of PFKFB3 and PFKFB4 provides sufficient glucose metabolism to satisfy the bioenergetics demand and redox homeostasis requirements of cancer cells. Various reversible post-translational modifications of PFKFB3 enable cancer cells to flexibly adapt glucose metabolism in response to diverse stress conditions. In addition to playing important roles in tumor cell glucose metabolism, PFKFB3 and PFKFB4 are widely involved in multiple biological processes, such as cell cycle regulation, autophagy, and transcriptional regulation in a non-glycolysis-dependent manner.
Collapse
Affiliation(s)
- Mei Yi
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, Hunan 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Department of Dermatology, Xiangya Hospital, The Central South University, Changsha, 410008, Hunan, China
| | - Yuanyuan Ban
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, Hunan 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yixin Tan
- Department of Dermatology, Second Xiangya Hospital, The Central South University, Hunan Key Laboratory of Medical Epigenetics, Changsha, 410011, Hunan, China
| | - Wei Xiong
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, Hunan 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Guiyuan Li
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, Hunan 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bo Xiang
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, Hunan 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
49
|
Souho T, Lamboni L, Xiao L, Yang G. Cancer hallmarks and malignancy features: Gateway for improved targeted drug delivery. Biotechnol Adv 2018; 36:1928-1945. [DOI: 10.1016/j.biotechadv.2018.08.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 07/22/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022]
|
50
|
Kashyap T, Pramanik KK, Nath N, Mishra P, Singh AK, Nagini S, Rana A, Mishra R. Crosstalk between Raf-MEK-ERK and PI3K-Akt-GSK3β signaling networks promotes chemoresistance, invasion/migration and stemness via expression of CD44 variants (v4 and v6) in oral cancer. Oral Oncol 2018; 86:234-243. [PMID: 30409306 DOI: 10.1016/j.oraloncology.2018.09.028] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/28/2018] [Accepted: 09/28/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND The cell-surface glycoprotein CD44 is an important oral cancer stem cell (OCSC) marker and plays significant role in oral squamous cell carcinoma (OSCC) aggressiveness, however, the regulation of CD44 is incompletely understood. METHODS In the present study, 145 fresh human OSCC tissue specimens, including 18 adjacent normal, 42 noninvasive (N0), 53 invasive tumor samples (N1-3) and 32 chemo-radiation resistant samples (RCRT), were included. The expression of CD44 standard (CD44s) and variants (CD44v4, CD44v6); the activation of pERK1/2, GSK3β, NICD (Notch) pathways; the cell viability; and the MMP-9/-2 activity were assessed using RT-PCR, immunohistochemistry, Western blotting, MTT assay and gelatin zymography. OSCC cell lines, including parental (SCC9/SCC4) and Cisplatin-resistant (CisR-SCC9/-SCC4) cells, were used. Knock down of CD44v4/CD44v6 (by siRNA) or inactivation of MAPK/PI3K pathways using specific PD98059/LY294002 was achieved for in vitro analysis of chemoresistance and invasion/migration. RESULTS Elevated CD44 variants were associated with overall OSCC progression, chemoresistance and invasion. Positive correlations were observed, mainly between the expression of CD44v4 and the activation of ERK1/2 causing chemoresistance, whereas CD44v6 expression and inactivation of GSK3β caused invasiveness of OSCC. Cisplatin resistant, CisR-SCC9/SCC4 cell lines showed OCSC properties. Inhibition of MEK/ERK1/2 by SMI or knock down (KD) of CD44v4 by siRNA reversed cisplatin-resistance, whereas blocking the PI3K/Akt/GSK3β pathway by SMI or KD of CD44v6 isoforms by respective siRNA diminished invasion/metastasis potential. CONCLUSION Collectively, our results demonstrated that CD44v4 expression is more linked with ERK1/2 activation and promote cisplatin resistance, whereas CD44v6 expression is associated primarily with PI3K/Akt/GSK3β activation and driving tumor invasion/migration.
Collapse
Affiliation(s)
- Tanushree Kashyap
- Centre for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi 835205, Jharkhand, India
| | - Kamdeo K Pramanik
- Centre for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi 835205, Jharkhand, India
| | - Nidhi Nath
- Centre for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi 835205, Jharkhand, India
| | - Prajna Mishra
- Centre for Applied Chemistry, School of Natural Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi 835205, Jharkhand, India
| | - Abhay K Singh
- Centre for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi 835205, Jharkhand, India
| | - Siddavaram Nagini
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar 608 002, Tamil Nadu, India
| | - Ajay Rana
- Division of Surgical Oncology, Department of Surgery, College of Medicine, The University of Illinois at Chicago 840 S. Wood Street, Suite 601 Clinical Sciences Building, MC 958, Chicago, IL 60612 USA
| | - Rajakishore Mishra
- Centre for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi 835205, Jharkhand, India.
| |
Collapse
|