51
|
Assis AJB, Santana BLDO, Gualberto ACM, Pittella-Silva F. Therapeutic applications of CRISPR/Cas9 mediated targeted gene editing in acute lymphoblastic leukemia: current perspectives, future challenges, and clinical implications. Front Pharmacol 2023; 14:1322937. [PMID: 38130408 PMCID: PMC10733529 DOI: 10.3389/fphar.2023.1322937] [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: 10/17/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Acute Lymphoblastic Leukemia (ALL) is the predominant hematological malignancy in pediatric populations, originating from B- or T-cell precursors within the bone marrow. The disease exhibits a high degree of heterogeneity, both at the molecular level and in terms of clinical presentation. A complex interplay between inherited and acquired genetic alterations contributes to disease pathogenesis, often resulting in the disruption of cellular functions integral to the leukemogenic process. The advent of CRISPR/Cas9 as a gene editing tool has revolutionized biological research, underscoring its potential to modify specific genomic loci implicated in cancer. Enhanced understanding of molecular alterations in ALL has facilitated significant advancements in therapeutic strategies. In this review, we scrutinize the application of CRISPR/Cas9 as a tool for identifying genetic targets to improve therapy, circumvent drug resistance, and facilitate CAR-T cell-based immunotherapy. Additionally, we discuss the challenges and future prospects of CRISPR/Cas9 applications in ALL.
Collapse
Affiliation(s)
| | | | | | - Fabio Pittella-Silva
- Laboratory of Molecular Pathology of Cancer, Faculty of Health Sciences and Medicine, University of Brasília, Brasília, Brazil
| |
Collapse
|
52
|
Torabidastgerdooei S, Roy ME, Annabi B. A molecular signature for the G6PC3/SLC37A2/SLC37A4 interactors in glioblastoma disease progression and in the acquisition of a brain cancer stem cell phenotype. Front Endocrinol (Lausanne) 2023; 14:1265698. [PMID: 38034009 PMCID: PMC10687460 DOI: 10.3389/fendo.2023.1265698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Background Glycogen plays an important role in glucose homeostasis and contributes to key functions related to brain cancer cell survival in glioblastoma multiforme (GBM) disease progression. Such adaptive molecular mechanism is dependent on the glycogenolytic pathway and intracellular glucose-6-phosphate (G6P) sensing by brain cancer cells residing within those highly hypoxic tumors. The involvement of components of the glucose-6-phosphatase (G6Pase) system remains however elusive. Objective We questioned the gene expression levels of components of the G6Pase system in GBM tissues and their functional impact in the control of the invasive and brain cancer stem cells (CSC) phenotypes. Methods In silico analysis of transcript levels in GBM tumor tissues was done by GEPIA. Total RNA was extracted and gene expression of G6PC1-3 as well as of SLC37A1-4 members analyzed by qPCR in four human brain cancer cell lines and from clinically annotated brain tumor cDNA arrays. Transient siRNA-mediated gene silencing was used to assess the impact of TGF-β-induced epithelial-to-mesenchymal transition (EMT) and cell chemotaxis. Three-dimensional (3D) neurosphere cultures were generated to recapitulate the brain CSC phenotype. Results Higher expression in G6PC3, SLC37A2, and SLC37A4 was found in GBM tumor tissues in comparison to low-grade glioma and healthy tissue. The expression of these genes was also found elevated in established human U87, U251, U118, and U138 GBM cell models compared to human HepG2 hepatoma cells. SLC37A4/G6PC3, but not SLC37A2, levels were induced in 3D CD133/SOX2-positive U87 neurospheres when compared to 2D monolayers. Silencing of SLC37A4/G6PC3 altered TGF-β-induced EMT biomarker SNAIL and cell chemotaxis. Conclusion Two members of the G6Pase system, G6PC3 and SLC37A4, associate with GBM disease progression and regulate the metabolic reprogramming of an invasive and CSC phenotype. Such molecular signature may support their role in cancer cell survival and chemoresistance and become future therapeutic targets.
Collapse
Affiliation(s)
| | | | - Borhane Annabi
- Laboratoire d’Oncologie Moléculaire, Centre de recherche CERMO-FC, Département de Chimie, Université du Québec à Montréal, Montreal, QC, Canada
| |
Collapse
|
53
|
Čižmáriková M, Michalková R, Mirossay L, Mojžišová G, Zigová M, Bardelčíková A, Mojžiš J. Ellagic Acid and Cancer Hallmarks: Insights from Experimental Evidence. Biomolecules 2023; 13:1653. [PMID: 38002335 PMCID: PMC10669545 DOI: 10.3390/biom13111653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is a complex and multifaceted disease with a high global incidence and mortality rate. Although cancer therapy has evolved significantly over the years, numerous challenges persist on the path to effectively combating this multifaceted disease. Natural compounds derived from plants, fungi, or marine organisms have garnered considerable attention as potential therapeutic agents in the field of cancer research. Ellagic acid (EA), a natural polyphenolic compound found in various fruits and nuts, has emerged as a potential cancer prevention and treatment agent. This review summarizes the experimental evidence supporting the role of EA in targeting key hallmarks of cancer, including proliferation, angiogenesis, apoptosis evasion, immune evasion, inflammation, genomic instability, and more. We discuss the molecular mechanisms by which EA modulates signaling pathways and molecular targets involved in these cancer hallmarks, based on in vitro and in vivo studies. The multifaceted actions of EA make it a promising candidate for cancer prevention and therapy. Understanding its impact on cancer biology can pave the way for developing novel strategies to combat this complex disease.
Collapse
Affiliation(s)
- Martina Čižmáriková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Radka Michalková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Gabriela Mojžišová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia;
| | - Martina Zigová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Annamária Bardelčíková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| |
Collapse
|
54
|
Cappello A, Tosetti G, Smirnov A, Ganini C, Yang X, Shi Y, Wang Y, Melino G, Bernassola F, Candi E. p63 orchestrates serine and one carbon metabolism enzymes expression in head and neck cancer. Biol Direct 2023; 18:73. [PMID: 37946250 PMCID: PMC10636826 DOI: 10.1186/s13062-023-00426-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is characterized by high proliferation and limited differentiation. The altered expression of the p53 family members, and specifically of p63, represents a pivotal event in the pathogenesis of HNSCC. Physiologically, p63 affects metabolism through the direct transactivation of the enzyme hexokinase 2, and subsequently controls the proliferation of epithelial cells; nonetheless, its role in cancer metabolism is still largely unclear. The high energetic demand of cancer and the consequent needs of a metabolic reshape, also involve the serine and glycine catabolic and anabolic pathways, including the one carbon metabolism (OCM), to produce energetic compounds (purines) and to maintain cellular homeostasis (glutathione and S-adenosylmethionine). RESULTS The involvement in serine/glycine starvation by other p53 family members has been reported, including HNSCC. Here, we show that in HNSCC p63 controls the expression of the enzymes regulating the serine biosynthesis and one carbon metabolism. p63 binds the promoter region of genes involved in the serine biosynthesis as well as in the one carbon metabolism. p63 silencing in a HNSCC cell line affects the mRNA and protein levels of these selected enzymes. Moreover, the higher expression of TP63 and its target enzymes, negatively impacts on the overall survival of HNSCC patients. CONCLUSION These data indicate a direct role of p63 in the metabolic regulation of HNSCC with significant clinical effects.
Collapse
Affiliation(s)
- Angela Cappello
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", 70121, Bari, Italy
| | - Giulia Tosetti
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Artem Smirnov
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, 00167, Rome, Italy
| | - Carlo Ganini
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", 70121, Bari, Italy
- Division of Medical Oncology, A.O.U. Policlinico di Bari, 70124, Bari, Italy
| | - Xue Yang
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
- Department of Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China
- National Center for Liver Cancer, Shanghai, 201805, China
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institute for Translational Medicine, Soochow University, Suzhou, China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Francesca Bernassola
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy.
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, 00167, Rome, Italy.
| |
Collapse
|
55
|
You S, Zhang J, Yu L, Li Z, Zhang J, Zhao N, Xie Z, Li Y, Akram Z, Sun S. Construction of SLC16A1/3 Targeted Gallic Acid-Iron-Embelin Nanoparticles for Regulating Glycolysis and Redox Pathways in Cervical Cancer. Mol Pharm 2023; 20:4574-4586. [PMID: 37307591 DOI: 10.1021/acs.molpharmaceut.3c00294] [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] [Indexed: 06/14/2023]
Abstract
SLC16A1 and SLC16A3 (SLC16A1/3) are highly expressed in cervical cancers and associated with the malignant biological behavior of cancer. SLC16A1/3 is the critical hub for regulating the internal and external environment, glycolysis, and redox homeostasis in cervical cancer cells. Inhibiting SLC16A1/3 provides a new thought to eliminate cervical cancer effectively. There are few reports on effective treatment strategies to eliminate cervical cancer by simultaneously targeting SLC16A1/3. GEO database analysis and quantitative reverse transcription polymerase chain reaction experiment were used to confirm the high expression of SLC16A1/3. The potential inhibitor of SLC16A1/3 was screened from Siwu Decoction by using network pharmacology and molecular docking technology. The mRNA levels and protein levels of SLC16A1/3 in SiHa and HeLa cells treated by Embelin (EMB) were clarified, respectively. Furthermore, the Gallic acid-iron (GA-Fe) drug delivery system was used to improve its anti-cancer performance. Compared with normal cervical cells, SLC16A1/3 mRNA was over-expressed in SiHa and HeLa cells. Through the analysis of Siwu Decoction, a simultaneously targeted SLC16A1/3 inhibitor EMB was discovered. It was found for the first time that EMB promoted lactic acid accumulation and further induced redox dyshomeostasis and glycolysis disorder by simultaneously inhibiting SLC16A1/3. The gallic acid-iron-Embelin (GA-Fe@EMB) drug delivery system delivered EMB, which had a synergistic anti-cervical cancer effect. Under the irradiation of a near-infrared laser, the GA-Fe@EMB could elevate the temperature of the tumor area effectively. Subsequently, EMB was released and mediated the lactic acid accumulation and the GA-Fe nanoparticle synergistic Fenton reaction to promote ROS accumulation, thereby increasing the lethality of the nanoparticles on cervical cancer cells. GA-Fe@EMB can target cervical cancer marker SLC16A1/3 to regulate glycolysis and redox pathways, synergistically with photothermal therapy, which provides a new avenue for the synergistic treatment of malignant cervical cancer.
Collapse
Affiliation(s)
- Shiwan You
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Pharmacy, Shihezi 832003, Xinjiang, China
| | - Jing Zhang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Pharmacy, Shihezi 832003, Xinjiang, China
- School of Medicine, Xinjiang University of Science & Technology, Korla, 841000, China
| | - Lan Yu
- Shanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Zuoping Li
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Pharmacy, Shihezi 832003, Xinjiang, China
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Chemistry and Chemical Engineering, Shihezi 832002, Xinjiang, China
| | - Jiaru Zhang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Pharmacy, Shihezi 832003, Xinjiang, China
| | - Na Zhao
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Pharmacy, Shihezi 832003, Xinjiang, China
| | - Zhenzhen Xie
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Pharmacy, Shihezi 832003, Xinjiang, China
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Chemistry and Chemical Engineering, Shihezi 832002, Xinjiang, China
| | - Youping Li
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Pharmacy, Shihezi 832003, Xinjiang, China
| | - Zubair Akram
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Chemistry and Chemical Engineering, Shihezi 832002, Xinjiang, China
| | - Shiguo Sun
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University College of Pharmacy, Shihezi 832003, Xinjiang, China
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| |
Collapse
|
56
|
Bahrani HMH, Ghobeh M, Homayouni Tabrizi M. The anticancer, anti-oxidant, and antibacterial activities of chitosan-lecithin-coated parthenolide/tyrosol hybrid nanoparticles. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:1603-1617. [PMID: 36755525 DOI: 10.1080/09205063.2023.2177473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/29/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Tyrosol (TYR) and parthenolide (PLT) have been used as synthetic antioxidant and natural anticancer compounds. In the current study, we aimed to synthesize an encapsulated complex of both PLT and TYR in a hybrid coating layer consisting of lecithin and chitosan molecules, a proper biocompatible drug delivery system to evaluate its antibacterial and anticancer potentials on human liver HepG2 and pancreatic Panc cancer cell lines. The chitosan-lecithin-coated PLT/TYR nanoparticles (clPT-NPs) were synthesized applying an auto-self-assembling method. The clPT-NPs were characterized utilizing DLS, FTIR, zeta potential, and TEM analysis. The clPT-NPs' antioxidant activity was measured by running ABTS and DPPH antioxidant assays. Moreover, the antibacterial potential of clPT-NPs was evaluated by applying disk diffusion, MIC, and MBC assays. Finally, the nanoparticles' cytotoxicity and apoptotic activity were studied by conducting MTT, Flow cytometry, AO/PI cell staining, and real-time PCR techniques. The clPT-NPs (38 nm) exhibited significant antioxidant activity by inhibiting ABTS and DPPH radicals at 187 and 290 μg/mL IC50 concentrations, respectively. Also, the nanoparticles induced a notable antibacterial activity against Staphylococcus aureus at 0.0625 mg/mL MIC and 0.125 mg/mL MBC concentrations. The clPT-NPs selectively decreased the cancer cells' survival and increased the apoptotic dead cells by up-regulating apoptotic gene expression (BAX and Cas-8) and down-regulating BCL-2 anti-apoptotic gene expression. The PLT toxicity has been merged with improved TYR antioxidant activity, which has been functionalized in a safe, biocompatible hybrid nano-delivery system.
Collapse
Affiliation(s)
| | - Maryam Ghobeh
- Department of Biology, Science and Research branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
57
|
Fan TWM, Winnike J, Al-Attar A, Belshoff AC, Lorkiewicz PK, Tan JL, Wu M, Higashi RM, Lane AN. Differential Inhibition of Anaplerotic Pyruvate Carboxylation and Glutaminolysis-Fueled Anabolism Underlies Distinct Toxicity of Selenium Agents in Human Lung Cancer. Metabolites 2023; 13:774. [PMID: 37512481 PMCID: PMC10383978 DOI: 10.3390/metabo13070774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023] Open
Abstract
Past chemopreventive human trials on dietary selenium supplements produced controversial outcomes. They largely employed selenomethionine (SeM)-based diets. SeM was less toxic than selenite or methylseleninic acid (MSeA) to lung cancer cells. We thus investigated the toxic action of these Se agents in two non-small cell lung cancer (NSCLC) cell lines and ex vivo organotypic cultures (OTC) of NSCLC patient lung tissues. Stable isotope-resolved metabolomics (SIRM) using 13C6-glucose and 13C5,15N2-glutamine tracers with gene knockdowns were employed to examine metabolic dysregulations associated with cell type- and treatment-dependent phenotypic changes. Inhibition of key anaplerotic processes, pyruvate carboxylation (PyC) and glutaminolysis were elicited by exposure to MSeA and selenite but not by SeM. They were accompanied by distinct anabolic dysregulation and reflected cell type-dependent changes in proliferation/death/cell cycle arrest. NSCLC OTC showed similar responses of PyC and/or glutaminolysis to the three agents, which correlated with tissue damages. Altogether, we found differential perturbations in anaplerosis-fueled anabolic pathways to underlie the distinct anti-cancer actions of the three Se agents, which could also explain the failure of SeM-based chemoprevention trials.
Collapse
Affiliation(s)
- Teresa W.-M. Fan
- Center for Environmental and Systems Biochemistry, Department Toxicology & Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA; (A.A.-A.); (R.M.H.); (A.N.L.)
| | - Jason Winnike
- Department of Chemistry, University of Louisville, Louisville, KY 40202, USA; (J.W.); (A.C.B.); (P.K.L.)
| | - Ahmad Al-Attar
- Center for Environmental and Systems Biochemistry, Department Toxicology & Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA; (A.A.-A.); (R.M.H.); (A.N.L.)
| | - Alexander C. Belshoff
- Department of Chemistry, University of Louisville, Louisville, KY 40202, USA; (J.W.); (A.C.B.); (P.K.L.)
| | - Pawel K. Lorkiewicz
- Department of Chemistry, University of Louisville, Louisville, KY 40202, USA; (J.W.); (A.C.B.); (P.K.L.)
| | - Jin Lian Tan
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Min Wu
- Seahorse Bioscience, Billerica, MA 01862, USA
| | - Richard M. Higashi
- Center for Environmental and Systems Biochemistry, Department Toxicology & Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA; (A.A.-A.); (R.M.H.); (A.N.L.)
| | - Andrew N. Lane
- Center for Environmental and Systems Biochemistry, Department Toxicology & Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA; (A.A.-A.); (R.M.H.); (A.N.L.)
| |
Collapse
|
58
|
Tambay V, Raymond VA, Goossens C, Rousseau L, Turcotte S, Bilodeau M. Metabolomics-Guided Identification of a Distinctive Hepatocellular Carcinoma Signature. Cancers (Basel) 2023; 15:3232. [PMID: 37370840 DOI: 10.3390/cancers15123232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a major contributor to cancer-related morbidity and mortality burdens globally. Given the fundamental metabolic activity of hepatocytes within the liver, hepatocarcinogenesis is bound to be characterized by alterations in metabolite profiles as a manifestation of metabolic reprogramming. METHODS HCC and adjacent non-tumoral liver specimens were obtained from patients after HCC resection. Global patterns in tissue metabolites were identified using non-targeted 1H Nuclear Magnetic Resonance (1H-NMR) spectroscopy whereas specific metabolites were quantified using targeted liquid chromatography-mass spectrometry (LC/MS). RESULTS Principal component analysis (PCA) within our 1H-NMR dataset identified a principal component (PC) one of 53.3%, along which the two sample groups were distinctively clustered. Univariate analysis of tissue specimens identified more than 150 metabolites significantly altered in HCC compared to non-tumoral liver. For LC/MS, PCA identified a PC1 of 45.2%, along which samples from HCC tissues and non-tumoral tissues were clearly separated. Supervised analysis (PLS-DA) identified decreases in tissue glutathione, succinate, glycerol-3-phosphate, alanine, malate, and AMP as the most important contributors to the metabolomic signature of HCC by LC/MS. CONCLUSIONS Together, 1H-NMR and LC/MS metabolomics have the capacity to distinguish HCC from non-tumoral liver. The characterization of such distinct profiles of metabolite abundances underscores the major metabolic alterations that result from hepatocarcinogenesis.
Collapse
Affiliation(s)
- Vincent Tambay
- Laboratoire d'Hépatologie Cellulaire, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X0A9, Canada
| | - Valérie-Ann Raymond
- Laboratoire d'Hépatologie Cellulaire, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X0A9, Canada
| | - Corentine Goossens
- Laboratoire d'Hépatologie Cellulaire, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X0A9, Canada
| | - Louise Rousseau
- Biobanque et Base de Données Hépatobiliaire et Pancréatique, Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X0C1, Canada
| | - Simon Turcotte
- Biobanque et Base de Données Hépatobiliaire et Pancréatique, Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X0C1, Canada
- Département de Chirurgie, Service de Transplantation Hépatique et de Chirurgie Hépatobiliaire et Pancréatique, Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X0C1, Canada
| | - Marc Bilodeau
- Laboratoire d'Hépatologie Cellulaire, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X0A9, Canada
- Département de Médecine, Université de Montréal, Montréal, QC H3T1J4, Canada
| |
Collapse
|
59
|
Bhoopathi P, Mannangatti P, Das SK, Fisher PB, Emdad L. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy. Adv Cancer Res 2023; 159:285-341. [PMID: 37268399 DOI: 10.1016/bs.acr.2023.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a prominent cause of cancer deaths worldwide, is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. More than 90% of patients with PDAC die within a year of being diagnosed. PDAC is increasing at a rate of 0.5-1.0% per year, and it is expected to be the second leading cause of cancer-related mortality by 2030. The resistance of tumor cells to chemotherapeutic drugs, which can be innate or acquired, is the primary factor contributing to the ineffectiveness of cancer treatments. Although many PDAC patients initially responds to standard of care (SOC) drugs they soon develop resistance caused partly by the substantial cellular heterogeneity seen in PDAC tissue and the tumor microenvironment (TME), which are considered key factors contributing to resistance to therapy. A deeper understanding of molecular mechanisms involved in PDAC progression and metastasis development, and the interplay of the TME in all these processes is essential to better comprehend the etiology and pathobiology of chemoresistance observed in PDAC. Recent research has recognized new therapeutic targets ushering in the development of innovative combinatorial therapies as well as enhancing our comprehension of several different cell death pathways. These approaches facilitate the lowering of the therapeutic threshold; however, the possibility of subsequent resistance development still remains a key issue and concern. Discoveries, that can target PDAC resistance, either alone or in combination, have the potential to serve as the foundation for future treatments that are effective without posing undue health risks. In this chapter, we discuss potential causes of PDAC chemoresistance and approaches for combating chemoresistance by targeting different pathways and different cellular functions associated with and mediating resistance.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
60
|
Nong S, Han X, Xiang Y, Qian Y, Wei Y, Zhang T, Tian K, Shen K, Yang J, Ma X. Metabolic reprogramming in cancer: Mechanisms and therapeutics. MedComm (Beijing) 2023; 4:e218. [PMID: 36994237 PMCID: PMC10041388 DOI: 10.1002/mco2.218] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 03/29/2023] Open
Abstract
Cancer cells characterized by uncontrolled growth and proliferation require altered metabolic processes to maintain this characteristic. Metabolic reprogramming is a process mediated by various factors, including oncogenes, tumor suppressor genes, changes in growth factors, and tumor-host cell interactions, which help to meet the needs of cancer cell anabolism and promote tumor development. Metabolic reprogramming in tumor cells is dynamically variable, depending on the tumor type and microenvironment, and reprogramming involves multiple metabolic pathways. These metabolic pathways have complex mechanisms and involve the coordination of various signaling molecules, proteins, and enzymes, which increases the resistance of tumor cells to traditional antitumor therapies. With the development of cancer therapies, metabolic reprogramming has been recognized as a new therapeutic target for metabolic changes in tumor cells. Therefore, understanding how multiple metabolic pathways in cancer cells change can provide a reference for the development of new therapies for tumor treatment. Here, we systemically reviewed the metabolic changes and their alteration factors, together with the current tumor regulation treatments and other possible treatments that are still under investigation. Continuous efforts are needed to further explore the mechanism of cancer metabolism reprogramming and corresponding metabolic treatments.
Collapse
Affiliation(s)
- Shiqi Nong
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Xiaoyue Han
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Yu Xiang
- Department of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Yuran Qian
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Yuhao Wei
- Department of Clinical MedicineWest China School of MedicineWest China HospitalSichuan UniversityChengduSichuanChina
| | - Tingyue Zhang
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Keyue Tian
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Kai Shen
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jing Yang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xuelei Ma
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
- Department of Biotherapy and Cancer CenterState Key Laboratory of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
61
|
Albosultan AI, Ghobeh M, Tabrizi MH. The Anticancer, Anti-metastatic, Anti-oxidant, and Anti-angiogenic Activity of Chitosan-coated Parthenolide/Bovine Serum Albumin Nanoparticles. J Inorg Organomet Polym Mater 2023. [DOI: 10.1007/s10904-023-02541-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
62
|
Shigeta K, Hasegawa M, Hishiki T, Naito Y, Baba Y, Mikami S, Matsumoto K, Mizuno R, Miyajima A, Kikuchi E, Saya H, Kosaka T, Oya M. IDH2 stabilizes HIF-1α-induced metabolic reprogramming and promotes chemoresistance in urothelial cancer. EMBO J 2023; 42:e110620. [PMID: 36637036 PMCID: PMC9929641 DOI: 10.15252/embj.2022110620] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 01/14/2023] Open
Abstract
Drug resistance contributes to poor therapeutic response in urothelial carcinoma (UC). Metabolomic analysis suggested metabolic reprogramming in gemcitabine-resistant urothelial carcinoma cells, whereby increased aerobic glycolysis and metabolic stimulation of the pentose phosphate pathway (PPP) promoted pyrimidine biosynthesis to increase the production of the gemcitabine competitor deoxycytidine triphosphate (dCTP) that diminishes its therapeutic effect. Furthermore, we observed that gain-of-function of isocitrate dehydrogenase 2 (IDH2) induced reductive glutamine metabolism to stabilize Hif-1α expression and consequently stimulate aerobic glycolysis and PPP bypass in gemcitabine-resistant UC cells. Interestingly, IDH2-mediated metabolic reprogramming also caused cross resistance to CDDP, by elevating the antioxidant defense via increased NADPH and glutathione production. Downregulation or pharmacological suppression of IDH2 restored chemosensitivity. Since the expression of key metabolic enzymes, such as TIGAR, TKT, and CTPS1, were affected by IDH2-mediated metabolic reprogramming and related to poor prognosis in patients, IDH2 might become a new therapeutic target for restoring chemosensitivity in chemo-resistant urothelial carcinoma.
Collapse
Affiliation(s)
- Keisuke Shigeta
- Department of UrologyKeio University School of MedicineTokyoJapan
| | | | - Takako Hishiki
- Department of Clinical and Translational Research centerKeio University School of MedicineTokyoJapan
- Department of BiochemistryKeio University School of MedicineTokyoJapan
| | - Yoshiko Naito
- Department of Clinical and Translational Research centerKeio University School of MedicineTokyoJapan
| | - Yuto Baba
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Shuji Mikami
- Division of PathologyKeio University School of MedicineTokyoJapan
| | | | - Ryuichi Mizuno
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Akira Miyajima
- Department of UrologyTokai University School of MedicineTokyoJapan
| | - Eiji Kikuchi
- Department of UrologyKeio University School of MedicineTokyoJapan
- Department of UrologySt. Marianna University School of MedicineKanagawaJapan
| | - Hideyuki Saya
- Department of Clinical and Translational Research centerKeio University School of MedicineTokyoJapan
- Division of Gene RegulationInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| | - Takeo Kosaka
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Mototsugu Oya
- Department of UrologyKeio University School of MedicineTokyoJapan
| |
Collapse
|
63
|
Abdel-Hamid MS, Mansour AM, Hassan MH, Abdelhady R, Elsadek BEM, El-Sayed ESM, Salama SA. Estrogen Attenuates Diethylnitrosamine-Induced Hepatocellular Carcinoma in Female Rats via Modulation of Estrogen Receptor/FASN/CD36/IL-6 Axis. Biol Pharm Bull 2023; 46:1558-1568. [PMID: 37914358 DOI: 10.1248/bpb.b23-00342] [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] [Indexed: 11/03/2023]
Abstract
This study was designed to evaluate the potential protective impact of estrogen and estrogen receptor against diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC) in rats. The levels of liver injury serum biomarkers, liver content of interleukin-6 (IL-6), relative liver weight and distortion of liver histological pictures were significantly increased in ovariectomized (OVX) rats and SHAM rats that received DEN alone and were further exaggerated when DEN was combined with fulvestrant (F) compared to non-DEN treated rats. The OVX rats showed higher insults than SHAM rats. The tapering impact on these parameters was clear in OVX rats that received estradiol benzoate (EB), silymarin (S) or orlistat (ORS). The immunohistochemistry and/or Western blot analysis of liver tissues showed a prominent increase in fatty acid synthase (FASN) and cluster of differentiation 36 (CD36) expressions in OVX and SHAM rats who received DEN and/ or F compared to SHAM rats. In contrast to S, treatment of OVX rats with EB mitigated DEN-induced expression of FASN and CD36 in liver tissue, while ORS improved DEN-induced expression of FASN. In conclusion, the protective effect against HCC was mediated via estrogen receptor alpha (ER-α) which abrogates its downstream genes involved in lipid metabolism namely FASN and CD36 depriving the tumor from survival vital energy source. In addition, ORS induced similar mitigating effect against DEN-induced HCC which could be attributed to FASN inhibition and anti-inflammatory effect. Furthermore, S alleviated DEN-induced HCC, independent of its estrogenic effect.
Collapse
Affiliation(s)
| | - Ahmed M Mansour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University
| | - Memy H Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University
| | - Rasha Abdelhady
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Fayoum University
| | - Bakheet E M Elsadek
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University
| | - El-Sayed M El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University
| | - Salama A Salama
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University
| |
Collapse
|
64
|
Liu X, Fang X, Lu L, Liu G. Prognostic significance and immune landscape of a fatty acid metabolism-related gene signature in colon adenocarcinoma. Front Genet 2022; 13:996625. [PMID: 36568396 PMCID: PMC9780302 DOI: 10.3389/fgene.2022.996625] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Background: Fatty acid metabolism (FAM), as a hallmark of caner, plays important roles in tumor initiation and carcinogenesis. However, the significance of fatty acid metabolism-related genes in colon adenocarcinoma (COAD) are largely unknown. Methods: RNA sequencing data and clinical information were downloaded from the Cancer Genome Atlas (TCGA) cohort. Univariate and multivariate Cox regression analyses were utilized to construct a fatty acid metabolism-related gene signature. Kaplan-Meier survival and receiver operating characteristic (ROC) analyses were used to verify the performance of this signature. GEO datasets were applied to validate the signature. Maftools package was utilized to analyze the mutation profiles of this signature. Correlation between the risk signature and stemness scores was compared by RNA stemness score (RNAss). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene set variation analysis (GSVA) were performed to explore the potential functions and signaling pathways. Immune landscape of the signature was explored by analyzing different immune cells infiltration, immune functions and microsatellite instability. A nomogram was constructed by combining the risk signature and multiple clinical factors. Expression levels and prognostic values of the risk genes were revealed in the cancer genome atlas and GEO databases. Moreover, the expression the risk genes were measured in cell lines using real time quantitative PCR (qRT-PCR). Results: Eight fatty acid metabolism-related genes (CD36, ENO3, MORC2, PTGR1, SUCLG2, ELOVL3, ELOVL6 and CPT2) were used to construct a risk signature. This signature demonstrated better prognostic value than other clinicopathological parameters, with AUC value was 0.734 according to the cancer genome atlas database. There was negative correlation between the riskscore and RNA stemness score. The patients in the high-risk group demonstrated higher infiltration of M0 macrophages, and less infiltration of activated CD4 memory T cells and Eosinophils. There were more MSI patients in the high-risk group than those in the low-risk group (38% vs. 30%). The risk scores of patients in the MSI group were slightly higher than those in the microsatellite stability group. Gene ontology, kyoto encyclopedia of genes and genomes and gene set variation analysis enrichment analyses showed that several metabolism-related functions and signaling pathways were enriched. A nomogram showed good predictive capability of the signature. Moreover, qRT-PCR revealed upregulated expression of ENO3, MORC2, SUCLG2 and ELOVL6, and downregulated expression of CPT2 in all examined colon adenocarcinoma cell lines. Conclusion: This study provided novel insights into a fatty acid metabolism-related signature in the prognosis an immune landscape of colon adenocarcinoma patients.
Collapse
Affiliation(s)
| | | | - Lin Lu
- *Correspondence: Guolong Liu, ; Lin Lu,
| | | |
Collapse
|