1
|
Jiang L, Huang M, Kwan KY, Wang Y, Ma X, Abo-Raya MH, Hu M. Stress Response of the Juvenile Tri-Spine Horseshoe Crab Tachypleus tridentatus to Hypoxia: Insights From Phenotypic, Metabolomic, and Microbial Analyses. Integr Zool 2025. [PMID: 39810487 DOI: 10.1111/1749-4877.12948] [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: 08/30/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Over the past few decades, ocean hypoxia has been increasing due to human activities. Hypoxic stress, characterized by a reduced level of dissolved oxygen, is an escalating threat to marine ecosystems, with potentially devastating effects on the viability of endangered species such as the tri-spine horseshoe crab Tachypleus tridentatus. Even though this species is remarkably resilient to low oxygen levels, persistent hypoxia can negatively impact its population's survivability. The objective of this research was to examine the impact of short-term hypoxia on the behavior, gut microbiota, and metabolomics of juvenile T. tridentatus. First instar juvenile horseshoe crabs were subjected to hypoxic stress (2 mg O2/L) for 14 days and then recovered for 7 days in an environment with normal dissolved oxygen. The findings demonstrated that short-term hypoxia reduced the rates of swimming and burrowing of horseshoe crabs, and induced the change of tissue metabolites and intestinal flora malfunction. Additionally, in the hypoxia groups on day 14, 86 distinct metabolites showed a trend of downregulation, while 29 metabolites showed an upregulation trend. Arginine biosynthesis; histidine metabolism; vitamin B6 metabolism; aminoacyl-tRNA biosynthesis; and alanine, aspartate, and glutamate metabolism were the top five metabolic KEGG pathways (p < 0.05) enriched with 8 metabolites. In conclusion, our results provided new insights related to the behaviors, microbiota, and metabolites involved in juvenile T. tridentatus exposed to short-term hypoxic conditions and confirmed that hypoxia impairs their behavioral and physiological status.
Collapse
Affiliation(s)
- Lingfeng Jiang
- Fisheries College of Jimei University, Xiamen, Fujian Province, China
- Key Laboratory of Tropical Marine Ecosystem and Bioresource, Fourth Institute of Oceanography, Ministry of Natural Resources, Beihai, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Meilian Huang
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Kit Yue Kwan
- Fisheries College of Jimei University, Xiamen, Fujian Province, China
| | - Youji Wang
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Xiaowan Ma
- Key Laboratory of Tropical Marine Ecosystem and Bioresource, Fourth Institute of Oceanography, Ministry of Natural Resources, Beihai, China
| | - Mohamed H Abo-Raya
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai, China
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, Egypt
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Menghong Hu
- Key Laboratory of Tropical Marine Ecosystem and Bioresource, Fourth Institute of Oceanography, Ministry of Natural Resources, Beihai, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai, China
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| |
Collapse
|
2
|
Pandey AR, Kumar A, Shrivastava NK, Singh J, Yadav S, Sonkar AB, Kumar D, Kumar R, Saeedan AS, Ansari MN, Aldossary SA, Akhter Y, Kaithwas G. Advancing siRNA Therapeutics targeting MCT-4: A Multifaceted approach integrating Arithmetical Designing, Screening, and molecular dynamics validation. Int Immunopharmacol 2025; 147:113980. [PMID: 39798472 DOI: 10.1016/j.intimp.2024.113980] [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: 06/19/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/15/2025]
Abstract
Monocarboxylate transporter 4 (MCT-4) is involved in various metabolic processes which are crucial in maintaining cellular pH and energy metabolism, and thus influence the tumor microenvironment. The study is aimed to rationally design effective Small interfering RNA (siRNA) that can silence MCT-4. We utilized a comprehensive workflow integrating multiple tools such as siDirect version 2.0, Oligowalk and i-score designer, to evaluate sequence features and predict target site accessibility, Guanine-Cytosine (GC) content and thermodynamic stability. Five (M1, M2, M3, M4 and M5) siRNAs sequences were retrived and subjected to further scrutiny on the account of off-target elimation, sequence conservation, secondary structure formation, and thermodynamic properties. The M1 demonstrated off targets and the M2 sequence showed secondry conformation and therefore M3, M4 and M5 were considered for further evaluation. Additionally, molecular docking and simulations (50 ns) were conducted with human Argonaute 2 protein (h-Arg-2). The post- molecular dynamics (MD) analysis revealed M4 (5'UUGAAGAAGACACUGACGG3') as a most appropriate siRNA candidate agsint MCT-4 on the basis of Root Mean Square Deviation (RMSD), Root Mean Square Fluctuation (RMSF), and H-Bond results. The Molecular Mechanics Poisson-Boltzmann Surface Area (MMPBSA) analysis was also performed to further validate the selected siRNA candidates, which further affirmed M4 (5'UUGAAGAAGACACUGACGG3') as an potential candidate for future in-vitro and in-vivo evaluation.
Collapse
Affiliation(s)
- Aadya Raj Pandey
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Anurag Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Neeraj Kumar Shrivastava
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Jyoti Singh
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Sneha Yadav
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Archana Bharti Sonkar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Dharmendra Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Rohit Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Abdulaziz S Saeedan
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Mohd Nazam Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Sara A Aldossary
- Department of Pharmaceutical Sciences, Clinical Pharmacy College, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Yusuf Akhter
- Department of Biotechnology, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road Lucknow, 226025, India
| | - Gaurav Kaithwas
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India.
| |
Collapse
|
3
|
Liu J, Feng C, Zhao R, Song H, Huang L, Jiang N, Yang X, Liu L, Duan C, Wan L, Gao Q, Sun L, Hou X, Liu M, Zhang Y, Zhang X, Zhang D, Wang Y, Li Y, Ma X, Zhong H, Min M, Wei C, Cao Y, Yang X. GP73 reinforces cytotoxic T-cell function by regulating HIF-1α and increasing antitumor efficacy. J Immunother Cancer 2025; 13:e009265. [PMID: 39762082 DOI: 10.1136/jitc-2024-009265] [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] [Accepted: 11/23/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Immunotherapy that targets immune checkpoints has achieved revolutionary success, but its application in solid tumors remains limited, highlighting the need for reliable enhancement of the efficacy of immunotherapy. Golgi protein 73 (GP73), a Golgi membrane protein, has been implicated in various cellular processes, including immune regulation. Recent studies suggested that GP73 may play a role in modulating the immune response in patients with cancer. In this study, we investigated the mechanism by which GP73 regulates T-cell-mediated antitumor immunity within the tumor microenvironment. METHODS We used T-cell specific GP73 knockout mice to establish MC38 and B16 tumor models to investigate the impact of GP73-deficient T cells on tumor growth. Single-cell sequencing was subsequently employed to classify tumor-infiltrating immune cells and assess changes in cytokines and metabolic genes. Through RNA sequencing, real-time quantitative PCR, western blotting, flow cytometry, seahorse analysis, glucose uptake, and L-lactic acid secretion assays, we explored how GP73 regulates hypoxia-inducible factor 1α (HIF-1α) to influence T-cell antitumor functionality. Furthermore, we established adoptive transfer experiments to study the ability of GP73-overexpressing T cells to combat tumors. Blood samples of patient with clinical tumor were collected to assess the relationship between immunotherapy efficacy and T-cell GP73 levels. RESULTS In this study, the absence of GP73 in mouse T cells promoted tumor growth and metastasis, accompanied by a decrease in the proportion of cytotoxic CD8+T cell subsets infiltrating the tumor and an increase in exhausted CD8+ T-cell subsets. Further analysis revealed that the effector function of CD8+T cells in tumors relies on glycolysis regulated by HIF-1α rather than immune checkpoints. GP73-deficient T cells exhibit severely impaired glycolysis in hypoxic environments, whereas ectopic GP73 expression restores HIF-1α levels. In adoptive immunotherapy, overexpression of GP73 in T cells inhibits tumor growth. In cytotoxicity assays, knockdown of GP73 affected the ability of CD8+T cells to kill target cells. Clinically, tumor immunotherapy partial response patients present significantly elevated levels of GP73 expression in T cells. CONCLUSIONS These findings reveal the role of GP73 in regulating T-cell glycolysis and may lead to new therapeutic strategies for the prognosis and treatment of clinical tumor immunotherapy.
Collapse
Affiliation(s)
- Jialong Liu
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
- Clinical Diagnosis Laboratory, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, Beijing, China
| | - Chao Feng
- Department of Basic Medical Sciences, 960th Hospital of People's Liberation Army Joint Logistic Support Force, Jinan, Shandong, China
- Department of Clinical Laboratory, The 969th Hospital of PLA, Hohhot, Inner Mongolia, China
| | - Ruzhou Zhao
- Department of Genetic Engineering, Academy of Military Medical Sciences, Beijing, Beijing, China
| | - Hongbin Song
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
| | - Linfei Huang
- Department of Genetic Engineering, Academy of Military Medical Sciences, Beijing, Beijing, China
| | - Nan Jiang
- Department of Pharmacy, PLA General Hospital Medical Supplies Center Department of Pharmacy, Beijing, Beijing, China
| | - Xiaopan Yang
- Department of Genetic Engineering, Academy of Military Medical Sciences, Beijing, Beijing, China
| | - Lanlan Liu
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
| | - Cuijuan Duan
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
| | - Luming Wan
- Department of Genetic Engineering, Academy of Military Medical Sciences, Beijing, Beijing, China
| | - Qi Gao
- Beijing Hotgen Biotech Co., Ltd, Beijing, Beijing, China
| | - Lijuan Sun
- Beijing Hotgen Biotech Co., Ltd, Beijing, Beijing, China
| | - Xufeng Hou
- Beijing Hotgen Biotech Co., Ltd, Beijing, Beijing, China
| | - Muyi Liu
- Department of Genetic Engineering, Academy of Military Medical Sciences, Beijing, Beijing, China
| | - Yanhong Zhang
- Department of Genetic Engineering, Academy of Military Medical Sciences, Beijing, Beijing, China
| | - Xuemiao Zhang
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
| | - Dandan Zhang
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
| | - Yufei Wang
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
| | - Yong Li
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xueping Ma
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
| | - Hui Zhong
- Beijing Hotgen Biotech Co., Ltd, Beijing, Beijing, China
| | - Min Min
- Department of Gastroenterlology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
| | - Congwen Wei
- Department of Genetic Engineering, Academy of Military Medical Sciences, Beijing, Beijing, China
| | - Yuan Cao
- Department of Basic Medical Sciences, 960th Hospital of People's Liberation Army Joint Logistic Support Force, Jinan, Shandong, China
| | - Xiaoli Yang
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, Beijing, China
| |
Collapse
|
4
|
Gamage CDB, Kim JH, Zhou R, Park SY, Pulat S, Varlı M, Nam SJ, Kim H. Plectalibertellenone A suppresses colorectal cancer cell motility and glucose metabolism by targeting TGF-β/Smad and Wnt pathways. Biofactors 2025; 51:e2120. [PMID: 39291722 DOI: 10.1002/biof.2120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/29/2024] [Indexed: 09/19/2024]
Abstract
Colorectal cancer (CRC) is the second most common cause of cancer-related death and represents a serious worldwide health problem. CRC metastasis decreases the survival rate of cancer patients, underscoring the need to identify novel anticancer agents and therapeutic targets. Here, we introduce Plectalibertellenone A (B) as a promising agent for the inhibition of CRC cell motility and glucose metabolism and explore its mechanism of action in CRC cells. Plectalibertellenone A suppressed TGF-β gene expression and the activation of the TGF-β/Smad signaling pathway, leading to reverse epithelial to mesenchymal transition (EMT) by modulating the expressions of EMT markers and transcriptional factors such as E-cadherin, N-cadherin, vimentin, Slug, Snail, Twist, and ZEB1/2. Furthermore, disruption of Wnt signaling inhibited CRC motility and glucose metabolism including glycolysis and oxidative phosphorylation, primarily affecting glycolytic enzymes, GLUT1, HK2, PKM2, LDHA, and HIF-1α under hypoxic condition. Therefore, Plectalibertellenone A is a potential drug candidate that can be developed into a promising anticancer treatment to prevent CRC metastasis and inhibit glucose metabolism.
Collapse
Affiliation(s)
| | - Jeong-Hyeon Kim
- Department of Chemistry and Nanoscience, Ewha Woman University, Seoul, Korea
| | - Rui Zhou
- College of Pharmacy, Sunchon National University, Sunchon, Jeonnam, Korea
| | - So-Yeon Park
- College of Pharmacy, Sunchon National University, Sunchon, Jeonnam, Korea
| | - Sultan Pulat
- College of Pharmacy, Sunchon National University, Sunchon, Jeonnam, Korea
| | - Mücahit Varlı
- College of Pharmacy, Sunchon National University, Sunchon, Jeonnam, Korea
| | - Sang-Jip Nam
- Department of Chemistry and Nanoscience, Ewha Woman University, Seoul, Korea
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, Sunchon, Jeonnam, Korea
| |
Collapse
|
5
|
Salnikova DI, Shchekotikhin AE, Scherbakov AM. [A commentary: New carbonic anhydrase IX-targeted probes for imaging hypoxic tumors]. Bull Cancer 2024:S0007-4551(24)00462-4. [PMID: 39690094 DOI: 10.1016/j.bulcan.2024.11.006] [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: 07/16/2024] [Revised: 10/30/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024]
Abstract
In 2023, the journals "Bioconjugate Chemistry" and "Sensors and Actuators B: Chemical" published two papers describing new biosensors for imaging hypoxic regions in tumors. Cao et al. combined acetazolamide (AZA) to target carbonic anhydrase IX (CA IX) with two tyrosine-derived Mn(II)-ethylenediaminetetraacetic acid chelates (TyEDTA) on a rigid triazine (TA) scaffold. The aim of this synthesis was to create a Mn(II)-based magnetic resonance imaging probe named AZA-TA-Mn. In a murine hypoxia model of esophageal squamous cell carcinoma, AZA-TA-Mn, at a low dose of 0.05mmol/kg, produced selective contrast enhancement over non-specific Gd-diethylenetriaminepentaacetic acid (0.1mmol/kg). A competition study involving the co-injection of free AZA and a Mn(II)-based magnetic resonance imaging probe confirmed the in vivo tumor selectivity of AZA-TA-Mn. Immunofluorescence staining of tissue sections confirmed the positive correlation between tumor accumulation of AZA-TA-Mn and overexpression of AC IX. Using CA IX as a biomarker of hypoxia, Cao et al. have thus illustrated a practical strategy for the development of novel imaging probes for specific regions of hypoxia in a tumor of interest.
Collapse
Affiliation(s)
- Diana Igorevna Salnikova
- Département de biologie expérimentale des tumeurs, institut de carcinogenèse, centre national de recherche médicale en oncologie N.N. Blokhin, ministère de la Santé de Russie, 24 Kashirskoe shosse, Moscou 115522, Fédération de Russie.
| | - Andrey Egorovich Shchekotikhin
- Laboratoire de transformation chimique des antibiotiques, institut Gause des nouveaux antibiotiques, 11, rue Bol'chaya Pirogovskaya, Moscou 119021, Fédération de Russie
| | - Alexander Mikhailovich Scherbakov
- Département de biologie expérimentale des tumeurs, institut de carcinogenèse, centre national de recherche médicale en oncologie N.N. Blokhin, ministère de la Santé de Russie, 24 Kashirskoe shosse, Moscou 115522, Fédération de Russie.
| |
Collapse
|
6
|
Altrawy A, Khalifa MM, Abdelmaksoud A, Khaled Y, Saleh ZM, Sobhy H, Abdel-Ghany S, Alqosaibi A, Al-Muhanna A, Almulhim J, El-Hashash A, Sabit H, Arneth B. Metabolites in the Dance: Deciphering Gut-Microbiota-Mediated Metabolic Reprogramming of the Breast Tumor Microenvironment. Cancers (Basel) 2024; 16:4132. [PMID: 39766032 PMCID: PMC11674667 DOI: 10.3390/cancers16244132] [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: 10/25/2024] [Revised: 11/29/2024] [Accepted: 12/08/2024] [Indexed: 01/05/2025] Open
Abstract
Breast cancer (BC), a major cause of death among women worldwide, has traditionally been linked to genetic and environmental factors. However, emerging research highlights the gut microbiome's significant role in shaping BC development, progression, and treatment outcomes. This review explores the intricate relationship between the gut microbiota and the breast tumor microenvironment, emphasizing how these microbes influence immune responses, inflammation, and metabolic pathways. Certain bacterial species in the gut either contribute to or hinder BC progression by producing metabolites that affect hormone metabolism, immune system pathways, and cellular signaling. An imbalance in gut bacteria, known as dysbiosis, has been associated with a heightened risk of BC, with metabolites like short-chain fatty acids (SCFAs) and enzymes such as β-glucuronidase playing key roles in this process. Additionally, the gut microbiota can impact the effectiveness of chemotherapy, as certain bacteria can degrade drugs like gemcitabine and irinotecan, leading to reduced treatment efficacy. Understanding the complex interactions between gut bacteria and BC may pave the way for innovative treatment approaches, including personalized microbiome-targeted therapies, such as probiotics and fecal microbiota transplants, offering new hope for more effective prevention, diagnosis, and treatment of BC.
Collapse
Affiliation(s)
- Afaf Altrawy
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt; (A.A.); (M.M.K.); (H.S.); (H.S.)
| | - Maye M. Khalifa
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt; (A.A.); (M.M.K.); (H.S.); (H.S.)
| | - Asmaa Abdelmaksoud
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt;
| | - Yomna Khaled
- Department of Bioinformatics and Functional Genomics, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt;
| | - Zeinab M. Saleh
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt;
| | - Hager Sobhy
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt; (A.A.); (M.M.K.); (H.S.); (H.S.)
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt;
| | - Amany Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Afnan Al-Muhanna
- King Fahad Hospital of the University, Alkhobar, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Jawaher Almulhim
- Department of Biological Sciences, King Faisal University, Alahsa 31982, Saudi Arabia;
| | - Ahmed El-Hashash
- Department of Biomedicine, Texas A&M University, College Station, TX 77840, USA;
| | - Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt; (A.A.); (M.M.K.); (H.S.); (H.S.)
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University, Feulgen Str., 35392 Giessen, Germany
| |
Collapse
|
7
|
Mir IA, Mir HA, Mehraj U, Bhat MY, Mir MA, Dar TA, Hussain MU. Chloroquine sensitises hypoxic colorectal cancer cells to ROS-mediated cell death via structural disruption of pyruvate dehydrogenase kinase 1. Free Radic Biol Med 2024; 227:656-666. [PMID: 39657842 DOI: 10.1016/j.freeradbiomed.2024.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Chloroquine (CQ), an autophagy antagonist, has been recently explored as a repurposable medicine for cancer; however the exact mechanism of its action is still not known. The present study investigated the effect of CQ on colorectal cancer cells to elucidate the underlying molecular mechanisms. We report for the first time that CQ suppresses hypoxia-induced growth and survival of HCT-116 cells by reducing glycolytic capacity and NAD+ production through inhibition of PDK1. Furthermore, in silico and in vitro studies revealed that CQ induces structural alteration in the PDK1 protein, leading to its destabilization and promotes its enhanced degradation by proteases. This degradation is in turn inhibited by the MG-132 protease inhibitor. Moreover, CQ-induced suppression of PDK1 results in mitochondrial damage through excessive production of ROS, as reflected by the reduction in mitochondrial membrane potential, which in turn triggers apoptosis through PARP cleavage and Caspase activation. These findings advocate CQ as a promising repurposable chemotherapeutic for colorectal cancer and a novel inhibitor of PDK1.
Collapse
Affiliation(s)
- Irfan Ahmad Mir
- Department of Biotechnology, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Hilal Ahmad Mir
- Department of Biotechnology, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India; Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Umar Mehraj
- Department of Pathology, Duke University, Durham, NC 27710, USA; Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, J&K, India
| | - Mohd Younus Bhat
- Department of Clinical Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, J&K, India
| | - Tanveer Ali Dar
- Department of Clinical Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Mahboob-Ul Hussain
- Department of Biotechnology, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India.
| |
Collapse
|
8
|
Youssef E, Zhao S, Purcell C, Olson GL, El-Deiry WS. Targeting the SMURF2-HIF1α axis: a new frontier in cancer therapy. Front Oncol 2024; 14:1484515. [PMID: 39697237 PMCID: PMC11652374 DOI: 10.3389/fonc.2024.1484515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024] Open
Abstract
The SMAD-specific E3 ubiquitin protein ligase 2 (SMURF2) has emerged as a critical regulator in cancer biology, modulating the stability of Hypoxia-Inducible Factor 1-alpha (HIF1α) and influencing a network of hypoxia-driven pathways within the tumor microenvironment (TME). SMURF2 targets HIF1α for ubiquitination and subsequent proteasomal degradation, disrupting hypoxic responses that promote cancer cell survival, metabolic reprogramming, angiogenesis, and resistance to therapy. Beyond its role in HIF1α regulation, SMURF2 exerts extensive control over cellular processes central to tumor progression, including chromatin remodeling, DNA damage repair, ferroptosis, and cellular stress responses. Notably, SMURF2's ability to promote ferroptotic cell death through GSTP1 degradation offers an alternative pathway to overcome apoptosis resistance, expanding therapeutic options for refractory cancers. This review delves into the multifaceted interactions between SMURF2 and HIF1α, emphasizing how their interplay impacts metabolic adaptations like the Warburg effect, immune evasion, and therapeutic resistance. We discuss SMURF2's dual functionality as both a tumor suppressor and, in certain contexts, an oncogenic factor, underscoring its potential as a highly versatile therapeutic target. Furthermore, modulating the SMURF2-HIF1α axis presents an innovative approach to destabilize hypoxia-dependent pathways, sensitizing tumors to chemotherapy, radiotherapy, and immune-based treatments. However, the complexity of SMURF2's interactions necessitate a thorough assessment of potential off-target effects and challenges in specificity, which must be addressed to optimize its clinical application. This review concludes by proposing future directions for research into the SMURF2-HIF1α pathway, aiming to refine targeted strategies that exploit this axis and address the adaptive mechanisms of aggressive tumors, ultimately advancing the landscape of precision oncology.
Collapse
Affiliation(s)
- Emile Youssef
- Research & Development, SMURF-Therapeutics, Inc., Providence, RI, United States
- Medical & Pharmacovigilance, Kapadi, Inc., Raleigh, NC, United States
| | - Shuai Zhao
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center at Brown University, Providence, RI, United States
| | - Connor Purcell
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center at Brown University, Providence, RI, United States
| | - Gary L. Olson
- Medicinal Chemistry & Drug Discovery, Provid Pharmaceuticals, Inc., Monmouth Junction, NJ, United States
| | - Wafik S. El-Deiry
- Research & Development, SMURF-Therapeutics, Inc., Providence, RI, United States
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center at Brown University, Providence, RI, United States
| |
Collapse
|
9
|
Wang Y, Xiao B, Li J, Zhang M, Zhang L, Chen L, Zhang J, Chen G, Zhang W. Hypoxia regulates small extracellular vesicle biogenesis and cargo sorting through HIF-1α/HRS signaling pathway in head and neck squamous cell carcinoma. Cell Signal 2024; 127:111546. [PMID: 39631619 DOI: 10.1016/j.cellsig.2024.111546] [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: 08/16/2024] [Revised: 10/28/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Small extracellular vesicles (sEVs) act as crucial messengers that transmit biological signals in hypoxic tumor microenvironment (TME), significantly impacting cancer progression. However, the precise mechanism by which hypoxia influences sEV biogenesis remains poorly understood. In this study, we observed increased sEV secretion and alterations in cargo composition in head and neck squamous cell carcinoma (HNSCC) cells under hypoxic conditions. We found that hepatocyte growth factor-regulated tyrosine kinase substrate (HRS), a key component of the endosomal sorting complexes required for transport (ESCRT), was upregulated during hypoxia. This upregulation activated the endosomal system and reduced degradation of multivesicular bodies (MVBs). HRS depletion altered the packaging of protein cargoes such as mitochondria-related proteins into sEVs under hypoxia, and these cargoes promoted a pro-tumorigenic phenotype of macrophages. Importantly, we demonstrated that HRS is transcriptionally activated by hypoxia inducible factor-1α (HIF-1α). Spatial transcriptomics and immunohistochemistry revealed a positive correlation between HRS and HIF-1α. These findings establish a link between the hypoxic response, sEV biogenesis, and cargo packaging, enhancing our understanding of how the hypoxic TME influences sEV biogenesis in HNSCC cells.
Collapse
Affiliation(s)
- Yiman Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Bolin Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jinbang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Mengyao Zhang
- Department of Thyroid and Breast Surgery, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Linzhou Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Liguo Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jing Zhang
- Department of Thyroid and Breast Surgery, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Wei Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
10
|
da Silva YB, Bedogni G, de Andrade Picanço G, de Souza JY, Nunes WS, da Costa TL, de Campos GB, Vargas Michelena L, Salomon CJ, Vinaud MC. Nanoformulated fenbendazole as an attractive approach for treating neurocysticercosis: in vitro and in vivo studies. Pharm Dev Technol 2024; 29:1093-1100. [PMID: 39508398 DOI: 10.1080/10837450.2024.2422936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE This work aimed to develop fenbendazole nanocrystals to evaluate their effects on the energetic metabolism of Taenia crassiceps cysticerci, following an intracranial inoculation in mice. METHODS Fenbendazole was nanoformulated by the antisolvent method using poloxamers 188 and 407 as stabilizers. The nanosuspensions were lyophilized without cryoprotectants and the nanocrystals were characterized in terms of particle size, zeta potential, and dissolution performance. The in vivo study was performed in infected animals treated with nanoformulated fenbendazole and raw drug and their metabolic impact was quantified by analyzing specific metabolites. RESULTS Fenbendazole samples were obtained by nanoprecipitation in > 80% yield. The average particle size of the freeze-dried samples was between 372 nm and 1600 nm. The nanosystems released a greater amount of the drug into the solution, compared to the raw drug. The in vivo studies showed that the fenbendazole-treated groups induced gluconeogenesis, partial blockage of the TCA cycle, and increased protein catabolism. As seen, the nanoformulation presented a greater effect on these parameters than the raw drug leading to remarkable modifications in the metabolism of the parasite, which in turn can influence the overall course of the infection and treatment outcomes. CONCLUSION These findings suggest that nanoformulated fenbendazole may be considered a valuable approach for an effective treatment of neurocysticercosis.
Collapse
Affiliation(s)
- Yngrid Batista da Silva
- Laboratory of Host-Parasite Relationship Studies, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goias, Brazil
| | - Giselle Bedogni
- Institute of Chemistry of Rosario (IQUIR-CONICET), Rosario, Argentina
| | | | - Jéssica Yonara de Souza
- Laboratory of Host-Parasite Relationship Studies, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goias, Brazil
| | - Waylla Silva Nunes
- Laboratory of Host-Parasite Relationship Studies, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goias, Brazil
| | - Tatiane Luiza da Costa
- Laboratory of Host-Parasite Relationship Studies, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goias, Brazil
| | - Geovana Batista de Campos
- Laboratory of Host-Parasite Relationship Studies, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goias, Brazil
| | | | - Claudio Javier Salomon
- Institute of Chemistry of Rosario (IQUIR-CONICET), Rosario, Argentina
- Faculty of Biochemical and Pharmaceutical Sciences, National University of Rosario, Rosario, Argentina
| | - Marina Clare Vinaud
- Laboratory of Host-Parasite Relationship Studies, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goias, Brazil
- Medicine Department, Biotechnology Institute, Federal University of Catalao, Goias, Brazil
| |
Collapse
|
11
|
Taghizadeh-Hesary F. Is Chronic Ice Water Ingestion a Risk Factor for Gastric Cancer Development? An Evidence-Based Hypothesis Focusing on East Asian Populations. Oncol Ther 2024; 12:629-646. [PMID: 39231856 PMCID: PMC11573998 DOI: 10.1007/s40487-024-00299-y] [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: 04/16/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
This article introduces a novel risk factor for gastric cancer (GC) by analyzing available epidemiological data from East Asian populations. A significantly higher age-standardized GC rate was observed in Japanese and Korean populations than in Chinese populations, despite nearly identical ethnicity, food habits, obesity rates, and alcohol consumption. Given the pivotal role of environmental factors in GC development, particularly for the intestinal type, a thorough evaluation of the lifestyles of these three populations was conducted to identify commonalities and disparities. It was observed that Japanese and Korean individuals prefer consuming ice water, while Chinese individuals tend to drink warm water, potentially influenced by traditional Chinese medicine disciplines. Considering the key features of GC development, a literature review was conducted to investigate the mechanisms through which the consumption of ice water might contribute to GC initiation and progression. Mechanistically, exposing gastric cells to hypothermia can increase the risk of carcinogenesis through multiple pathways. This includes the promotion of Helicobacter pylori colonization, prolonged gastric inflammation, and mitochondrial dysfunction in gastric cells. Furthermore, drinking ice water can enhance the survival, proliferation, and invasion of GC cells by releasing cold shock proteins, increasing gastric acid secretion, and delaying gastric emptying. Additionally, hypothermia can boost the immune evasion of cancer cells by weakening the antitumor immune system and activating different components of the tumor microenvironment. This paper also explores the association between exposure of GC cells to hypothermia and current insights into cancer hallmarks. These findings may partially elucidate the higher incidence of GC in Japanese and Korean populations and provide a clue for future experimental studies.Graphical abstract available for this article.
Collapse
Affiliation(s)
- Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Radiation Oncology Department, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Ma XN, Ho YK, Goie JYG, Ma CX, Sun ZB, Yao LQ, Zhu XL, Woo JY, Too HP, Li X. Evaluating the potential of off-the-shelf engineered mesenchymal stem cells for targeted Hepatocellular Carcinoma treatment: A multisite proof-of-concept study. Biomed Pharmacother 2024; 181:117676. [PMID: 39522266 DOI: 10.1016/j.biopha.2024.117676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Although combining 5-fluorouracil (5-FU) and Interferon-beta (IFNb) improves response rates in Hepatocellular Carcinoma (HCC), the outcomes remain suboptimal. This study investigates the feasibility of using highly transfected Mesenchymal Stem Cells (MSCs) to deliver a chemotherapeutic (5-FU) and an immunomodulator (IFNb) for localized HCC treatment. Considering the crucial role of cold-chain transportation in off-the-shelf allogeneic therapy, the study also assesses the quality and efficacy of frozen-thawed engineered MSCs, simulating a multisite study process. The engineered MSCs maintained their phenotypes and tumour tropism. With just 10 % engineered MSCs, a killing efficiency of over 70 % was achieved in Huh-7 and HepG2 cell lines in vitro. Coculture studies, soft agar assays, and in vivo experiments confirmed that MSCs are neither tumorigenic nor tumour-promoting. Tumour mass growth was inhibited by >80 % in the treated mice group. TUNEL, Annexin-V, and Ki67 staining confirmed DNA damage, cell death, and proliferation inhibition post-treatment. Blood chemistry and the weight of the mice were comparable to the control group, indicating a good safety profile. This proof-of-concept study demonstrates the efficacy and safety of off-the-shelf CDUPRT-IFNβ_MSCs in targeting hepatocellular carcinoma (HCC) growth. Evaluating the complete value chain of MSC therapy in early-stage preclinical studies is essential for justifying further investigation and clinical translation of this cell product.
Collapse
Affiliation(s)
- Xiao Ni Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China; Medicine Laboratory Centre, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yoon Khei Ho
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; AGeM Bio, Singapore 119276, Singapore; Singapore Innovate, Singapore 059911, Singapore
| | - Jian Yi Gerald Goie
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Cheng-Xu Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China; Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zong-Bin Sun
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Li-Qiong Yao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China; Medicine Laboratory Centre, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiao Liang Zhu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jun Yung Woo
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Heng-Phon Too
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, Gansu, China.
| |
Collapse
|
13
|
Duggal I, Kim J, Zhang Y, Wang J, Lu A, Maniruzzaman M. Additive manufacturing to fight cancer: Current Applications and Future Directions. Drug Discov Today 2024; 29:104218. [PMID: 39613181 DOI: 10.1016/j.drudis.2024.104218] [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: 06/17/2024] [Revised: 10/01/2024] [Accepted: 10/24/2024] [Indexed: 12/01/2024]
Abstract
3D printing has emerged as a powerful tool demonstrating effectiveness in early screening and targeted delivery for various types of tumors. Although the applications of additive manufacturing for cancer are widespread, the issues of scaling up, quality control and specificity remain. This review presents a comprehensive analysis of the current landscape of use of additive manufacturing in cancer diagnostics and treatment. Furthermore, it proceeds to elucidate the prominent current and future applications of 4D- and 5D-printed micro-swimmers in cancer treatment with particular emphasis on the significant progress made in magnetic, biological and light-based propulsion microrobots. Lastly, the current limitations and future research directions are enumerated. In summary, this paper serves as a comprehensive exploration of the remarkable contributions of additive manufacturing to the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Ishaan Duggal
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, A1920, Austin, TX 78712, USA
| | - Joon Kim
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, A1920, Austin, TX 78712, USA
| | - Yu Zhang
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, A1920, Austin, TX 78712, USA; PharmE3D Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Jiawei Wang
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, A1920, Austin, TX 78712, USA
| | - Anqi Lu
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, A1920, Austin, TX 78712, USA
| | - Mohammed Maniruzzaman
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, A1920, Austin, TX 78712, USA; PharmE3D Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA.
| |
Collapse
|
14
|
Kuduvalli SS, Senthilathiban DP, Biswas I, Antony JS, Subramani M, Anitha TS. The synergistic anti-Warburg efficacy of temozolomide, metformin and epigallocatechin gallate in glioblastoma. Toxicol Appl Pharmacol 2024; 493:117146. [PMID: 39510432 DOI: 10.1016/j.taap.2024.117146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/16/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024]
Abstract
An important hallmark of glioblastoma aggressiveness is its altered metabolism of glucose. This metabolic shift wherein the tumor cells employ aerobic glycolysis regardless of oxygen availability via reprogramming of mitochondrial oxidative phosphorylation is known as the Warburg effect. Previous literatures have linked this metabolic reprograming to tumor progression and glioblastoma cell proliferation making it a key target for targeted drug therapy. Based on this lacuna, the current study aimed to explore the therapeutic efficacy of the triple-drug combination of temozolomide, metformin and epigallocatechin gallate in attenuating Warburg effect and glucose uptake in glioblastoma both in vitro and in vivo. Our results showed that the triple-drug combination had significantly reduced glucose uptake and reversed the Warburg effect in glioblastoma cells and in the glioma-induced xenograft rat model. Thus, the triple-drug combination would serve as an effective therapeutic regime to hamper glioblastoma progression via altering glucose metabolism and improving the overall prognosis in patient setting.
Collapse
Affiliation(s)
- Shreyas S Kuduvalli
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry 607 402, India; Department of Microbial Biotechnology and Protein Research Laboratory, Institute of Advanced Studies in Science and Technology, Guwahati, Assam 781035, India.
| | - Daisy Precilla Senthilathiban
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry 607 402, India.
| | - Indrani Biswas
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry 607 402, India.
| | - Justin S Antony
- University Children's Hospital Tübingen, Department of General Pediatrics, Hematology /Oncology, Tübingen, Germany.
| | - Madhu Subramani
- ScirosBio, Al Khatem Tower, ADGM Square, Abu Dhabi, United Arab Emirates.
| | - T S Anitha
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605 014, India.
| |
Collapse
|
15
|
Sharma R, Kashyap M, Zayed H, Krishnia L, Kashyap MK. Artificial blood-hope and the challenges to combat tumor hypoxia for anti-cancer therapy. Med Biol Eng Comput 2024:10.1007/s11517-024-03233-6. [PMID: 39614063 DOI: 10.1007/s11517-024-03233-6] [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: 03/13/2024] [Accepted: 10/22/2024] [Indexed: 12/01/2024]
Abstract
The blood plays a vital role in the human body and serves as an intermediary between various physiological systems and organs. White blood cells, which are a part of the immune system, defend against infections and regulate the body temperature and pH balance. Blood platelets play a crucial role in clotting, the prevention of excessive bleeding, and the promotion of healing. Blood also serves as a courier system that transports hormones to facilitate communication and synchronization between different organs and systems in the body. The circulatory system, comprised of arteries, veins, and capillaries, plays a crucial role in the efficient transportation and connection of vital nutrients and oxygen. Despite the importance of natural blood, there are often supply shortages, compatibility issues, and medical conditions, which make alternatives such as artificial blood necessary. This is particularly relevant in cancer treatment, which was the focus of our study. In this study, we investigated the potential of artificial blood in cancer therapy, specifically to address tumor hypoxia. We also examined the potential of red blood cell substitutes such as hemoglobin-based oxygen carriers and perfluorocarbons. Additionally, we examined the production of hemoglobin using E. coli and the role of hemoglobin in oncogenesis. Furthermore, we explored the potential use of artificial platelets for cancer treatment. Our study emphasizes the significance of artificial blood in improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Rishabh Sharma
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Amity Education Valley, Panchgaon, Manesar (Gurugram), Haryana, 122413, India
| | - Manju Kashyap
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Amity Education Valley, Panchgaon, Manesar (Gurugram), Haryana, 122413, India
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Lucky Krishnia
- Amity Institute of Nanotechnology, Amity School of Applied Sciences, Amity University Haryana, Panchgaon, Manesar (Gurugram), Haryana, 122413, India.
| | - Manoj Kumar Kashyap
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Amity Education Valley, Panchgaon, Manesar (Gurugram), Haryana, 122413, India.
| |
Collapse
|
16
|
Mi L, Zhang H. Myriad factors and pathways influencing tumor radiotherapy resistance. Open Life Sci 2024; 19:20220992. [PMID: 39655194 PMCID: PMC11627069 DOI: 10.1515/biol-2022-0992] [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: 05/07/2024] [Revised: 09/09/2024] [Accepted: 09/30/2024] [Indexed: 12/12/2024] Open
Abstract
Radiotherapy is a cornerstone in the treatment of various tumors, yet radioresistance often leads to treatment failure and tumor recurrence. Several factors contribute to this resistance, including hypoxia, DNA repair mechanisms, and cancer stem cells. This review explores the diverse elements that drive tumor radiotherapy resistance. Historically, resistance has been attributed to cellular repair and tumor repopulation, but recent research has expanded this understanding. The tumor microenvironment - characterized by hypoxia, immune evasion, and stromal interactions - further complicates treatment. Additionally, molecular mechanisms such as aberrant signaling pathways, epigenetic modifications, and non-B-DNA structures play significant roles in mediating resistance. This review synthesizes current knowledge, highlighting the interplay of these factors and their clinical implications. Understanding these mechanisms is crucial for developing strategies to overcome resistance and improve therapeutic outcomes in cancer patients.
Collapse
Affiliation(s)
- Lanjuan Mi
- School of Life and Health Sciences, Huzhou College, Hu Zhou, China
| | - Hongquan Zhang
- The First Affiliated Hospital of Huzhou University, Hu Zhou, China
| |
Collapse
|
17
|
Miller C, Klyuzhin I, Chaussé G, Brosch-Lenz J, Koniar H, Shi K, Rahmim A, Uribe C. Impact of cell geometry, cellular uptake region, and tumour morphology on 225Ac and 177Lu dose distributions in prostate cancer. EJNMMI Phys 2024; 11:97. [PMID: 39570450 PMCID: PMC11582247 DOI: 10.1186/s40658-024-00700-9] [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: 06/03/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Radiopharmaceutical therapy with 225Ac- and 177Lu-PSMA has shown promising results for the treatment of prostate cancer. However, the distinct physical properties of alpha and beta radiation elicit varying cellular responses, which could be influenced by factors such as tumour morphology. In this study, we use simulations to examine how cell geometry, region of pharmaceutical uptake within the cell to model different internalization fractions, and the presence of tumour hypoxia and necrosis impact nucleus absorbed doses and dose heterogeneity with 225Ac and 177Lu. We also develop nucleus absorbed dose kernels for application to autoradiography images. METHODS We used the GATE Monte Carlo software to simulate three geometries of LNCaP prostate cancer cells (spherical, cubic, and ovoid) with activity of 225Ac or 177Lu internalized in the cytoplasm or bound to the extracellular membrane. Nucleus S-values were calculated for each geometry, source region, and isotope. The cell models were used to create nucleus absorbed dose kernels for each source region describing the dose to each nucleus in a cell layer, which were applied to simulated tumours composed of normoxic, hypoxic, or necrotic cancer cells to obtain dose rate maps. Absorbed doses within the tumours and dose heterogeneity were analyzed for each tumour morphology and isotope. Cell geometry made a minimal impact on S-values to the nucleus, however internalization resulted in higher nucleus doses. Applying the kernels to the simulated tumour maps showed that doses to each cell type varied between 225Ac and 177Lu depending on tumour morphology. Dose heterogeneity within tumours was slightly higher with 225Ac, however the tumour morphology made a larger impact on dose heterogeneity compared to the choice of isotope, with hypoxic and necrotic tumours having very heterogeneous dose distributions. CONCLUSIONS Cell geometry simplifications may still allow robust results in simulation studies. Furthermore, the morphology of the tumour itself may make a larger impact on treatment response compared to other variables such as ratio of internalization. Finally, nucleus absorbed dose kernels were created that could enable microdosimetric studies with autoradiography.
Collapse
Affiliation(s)
- Cassandra Miller
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Physics, University of British Columbia, Vancouver, BC, Canada
| | - Ivan Klyuzhin
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | | | - Julia Brosch-Lenz
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Helena Koniar
- Department of Physics, University of British Columbia, Vancouver, BC, Canada
| | - Kuangyu Shi
- Department of Nuclear Medicine, Bern University Hospital, The University of Bern, Bern, Switzerland
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Physics, University of British Columbia, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Carlos Uribe
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada.
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada.
- Molecular Imaging and Therapy, BC Cancer Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
18
|
Chen S, Liu J, He G, Tang N, Zeng Y. Research Hotspots and Trends in Global Cancer immunometabolism:A Bibliometric Analysis from 2000 to 2023. J Multidiscip Healthc 2024; 17:5117-5137. [PMID: 39553266 PMCID: PMC11568773 DOI: 10.2147/jmdh.s495330] [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: 09/18/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Background Cancer poses a major global health challenge, and immunotherapy, known as the third revolution in cancer treatment, has brought new hope to patients. The emerging field of immunometabolism has further enhanced the safety and efficacy of immunotherapy. Over the past two decades, this field has rapidly evolved in oncology, leading to numerous significant findings. This review systematically examines the literature on immunometabolism in cancer, visualizing research trends and identifying future directions. Methods A comprehensive literature search was conducted in the Web of Science, PubMed, and Scopus databases, covering publications from January 2000 to December 2023. We employed tools like Citespace, VOSviewer, and RStudio for visual analysis of publication trends, regional contributions, institutions, authors, journals, and keywords. Results A total of 3320 articles were published by 8090 authors across 1738 institutions, involving 71 countries. Leading contributors were China (n=469), the United States (n=361), and Germany (n=82). Harvard University was the most influential institution, while Frontiers in Immunology had the highest number of publications. The top research areas included glucose, lipid, and amino acid metabolism, the tumor microenvironment, and immune cell regulation. Conclusion International collaboration and interdisciplinary efforts are advancing the field of cancer immunometabolism. Future research will likely focus on the interplay between metabolism and immunity, metabolic markers, immune cell reprogramming, and tumor-immune metabolic competition.
Collapse
Affiliation(s)
- Shupeng Chen
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Jie Liu
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Guilian He
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Nana Tang
- Hematology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Yingjian Zeng
- Hematology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| |
Collapse
|
19
|
Ledda M, Pluchino A, Ragusa M. Exploring the Role of Genetic and Environmental Features in Colorectal Cancer Development: An Agent-Based Approach. ENTROPY (BASEL, SWITZERLAND) 2024; 26:923. [PMID: 39593869 PMCID: PMC11593013 DOI: 10.3390/e26110923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024]
Abstract
The complexity of issues in cancer research has led to the introduction of powerful computational tools to help experimental in vivo and in vitro methods. These tools, which typically focus on studying cell behavior and dynamic cell populations, range from systems of differential equations that are solved numerically to lattice models and agent-based simulations. In particular, agent-based models (ABMs) are increasingly used due to their ability to incorporate multi-scale features, ranging from the individual to the population level. This approach allows for the combination of statistically aggregated assumptions with individual heterogeneity. In this work, we present an ABM that simulates tumor progression in a colonic crypt, to provide an experimental in silico environment for testing results achieved in traditional laboratory research and developing alternative scenarios of tumor development. The model also allows some speculations about causal relationships in biologically inspired systems.
Collapse
Affiliation(s)
- Marco Ledda
- Dipartimento di Fisica e Astronomia Ettore Majorana, Università di Catania, 95123 Catania, Italy;
| | - Alessandro Pluchino
- Dipartimento di Fisica e Astronomia Ettore Majorana, Università di Catania, 95123 Catania, Italy;
- INFN Sezione di Catania, 95123 Catania, Italy
| | - Marco Ragusa
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica, Università di Catania, 95123 Catania, Italy;
| |
Collapse
|
20
|
Zhang T, Chen L, Kueth G, Shao E, Wang X, Ha T, Williams DL, Li C, Fan M, Yang K. Lactate's impact on immune cells in sepsis: unraveling the complex interplay. Front Immunol 2024; 15:1483400. [PMID: 39372401 PMCID: PMC11449721 DOI: 10.3389/fimmu.2024.1483400] [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] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
Lactate significantly impacts immune cell function in sepsis and septic shock, transcending its traditional view as just a metabolic byproduct. This review summarizes the role of lactate as a biomarker and its influence on immune cell dynamics, emphasizing its critical role in modulating immune responses during sepsis. Mechanistically, key lactate transporters like MCT1, MCT4, and the receptor GPR81 are crucial in mediating these effects. HIF-1α also plays a significant role in lactate-driven immune modulation. Additionally, lactate affects immune cell function through post-translational modifications such as lactylation, acetylation, and phosphorylation, which alter enzyme activities and protein functions. These interactions between lactate and immune cells are central to understanding sepsis-associated immune dysregulation, offering insights that can guide future research and improve therapeutic strategies to enhance patient outcomes.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Linjian Chen
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Gatkek Kueth
- James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Emily Shao
- Program in Neuroscience, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
| | - Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Tuanzhu Ha
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - David L. Williams
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Chuanfu Li
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Min Fan
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Kun Yang
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
21
|
Surguta SE, Baranyi M, Svajda L, Cserepes M, Ranđelović I, Tátrai E, Hegedűs B, Tóvári J. Differential effects of hypoxia on motility using various in vitro models of lung adenocarcinoma. Sci Rep 2024; 14:20482. [PMID: 39227650 PMCID: PMC11372077 DOI: 10.1038/s41598-024-70769-w] [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: 02/20/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related death globally. Metastasis is the most common reason of mortality in which hypoxia is suggested to have a pivotal role. However, the effect of hypoxia on the metastatic potential and migratory activity of cancer cells is largely unexplored and warrants detailed scientific investigations. Accordingly, we analyzed changes on cell proliferation and migratory activity both in single-cell migration and invasion under normoxic and hypoxic conditions in lung adenocarcinoma cell lines. Alterations in crucial genes and proteins associated with cellular response to hypoxia, epithelial-mesenchymal transition, proliferation and apoptosis were also analyzed. Generally, we observed no change in proliferation upon hypoxic conditions and no detectable induction of apoptosis. Interestingly, we observed that single-cell motility was generally reduced while invasion under confluent conditions using scratch assay was enhanced by hypoxia in most of the cell lines. Furthermore, we detected changes in the expression of EMT markers that are consistent with enhanced motility and metastasis-promoting effect of hypoxia. In summary, our study indicated cell line-, time of exposure- and migrational type-dependent effects of hypoxia in cellular proliferation, motility and gene expression. Our results contribute to better understanding and tackling cancer metastasis.
Collapse
Affiliation(s)
- Sára Eszter Surguta
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary.
- School of Ph.D. Studies, Semmelweis University, Budapest, 1085, Hungary.
| | - Marcell Baranyi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, 1091, Hungary
| | - Laura Svajda
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
- School of Ph.D. Studies, Semmelweis University, Budapest, 1085, Hungary
| | - Mihály Cserepes
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Ivan Ranđelović
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Enikő Tátrai
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Balázs Hegedűs
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, 1091, Hungary
- Department of Thoracic Surgery, University Medicine Essen - Ruhrlandklinik, University Duisburg-Essen, 45239, Essen, Germany
| | - József Tóvári
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
- School of Ph.D. Studies, Semmelweis University, Budapest, 1085, Hungary
| |
Collapse
|
22
|
Han P, Zhang B, Li Y, Gao R, Li X, Ren H, Shi P, Zhao A, Xue J, Yang A, Liang Y. MiR-183-5p inhibits lung squamous cell carcinoma survival through disrupting hypoxia adaptation mediated by HIF-1α/NDUFA4L2 axis. Oncogene 2024; 43:2821-2834. [PMID: 39154121 DOI: 10.1038/s41388-024-03129-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024]
Abstract
Hypoxia is a common feature of lung squamous cell carcinoma (LUSC), and hypoxia-inducible factor-1 (HIF-1) overexpression is associated with poor clinical outcome in LUSC. NADH dehydrogenase 1 alpha subcomplex subunit 4-like 2 (NDUFA4L2) is a recently identified target of HIF-1, but its roles in LUSC remain unclear. Herein, the expression and regulatory mechanisms of NDUFA4L2 were investigated in LUSC, and the influences on LUSC cell oxidative metabolism and survival of NDUFA4L2 were determined. The potential microRNA targeting to NDUFA4L2 was identified and its roles on LUSC cell were detected. We found that NDUFA4L2 were overexpressed in LUSC tissues, and that NDUFA4L2 expression correlated with shorter overall survival. NDUFA4L2 was regulated by HIF-1α under hypoxia, and NDUFA4L2 decreased mitochondrial reactive oxygen species (mitoROS) production through inhibiting mitochondrial complex I activity in LUSC cells. NDUFA4L2 silencing effectively suppressed LUSC cell growth and enhanced apoptosis by inducing mitoROS accumulation. Additionally, NDUFA4L2 was a target for miR-183-5p, and LUSC patients with high miR-183-5p levels had better prognoses. MiR-183-5p significantly induced mitoROS production and suppressed LUSC survival through negatively regulating NDUFA4L2 in vitro and in vivo. Our results suggested that regulation of NDUFA4L2 by HIF-1α is an important mechanism promoting LUSC progression under hypoxia. NDUFA4L2 inhibition using enforced miR-183-5p expression might be an effective strategy for LUSC treatment.
Collapse
Affiliation(s)
- Peng Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Boxiang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yixing Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Rui Gao
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xinru Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hui Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Puyu Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Aomei Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jianjun Xue
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Aimin Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yiqian Liang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
23
|
Taghizadeh-Hesary F, Ghadyani M, Kashanchi F, Behnam B. Exploring TSGA10 Function: A Crosstalk or Controlling Mechanism in the Signaling Pathway of Carcinogenesis? Cancers (Basel) 2024; 16:3044. [PMID: 39272902 PMCID: PMC11393850 DOI: 10.3390/cancers16173044] [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: 07/08/2024] [Revised: 08/18/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Cancer-specific antigens have been a significant area of focus in cancer treatment since their discovery in the mid-twentieth century. Cancer germline antigens are a class of antigens specifically overexpressed in germline tissues and cancer cells. Among these, TSGA10 (testis-specific gene antigen 10) is of great interest because of its crucial impact on cancer progression. Early studies explored TSGA10 expression in a variety of cancer types. More recent studies revealed that TSGA10 can suppress tumor progression by blocking cancer cell metabolism, angiogenesis, and metastasis. An open question regarding the TSGA10 is why cancer cells must express a protein that prevents their progression. To answer this question, we conducted a comprehensive review to engage the TSGA10 in the context of the current understanding of "malignant transformation". This review demonstrated that TSGA10 expression level in cancer cells depends on the cancer stage across malignant transformation. In addition, we evaluated how TSGA10 expression can prevent the "cancer hallmarks". Given this information, TSGA10 can be of great interest in developing effective targeted anti-cancer therapies.
Collapse
Affiliation(s)
- Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Mobina Ghadyani
- Chester Medical School, University of Chester, Chester CH2 1BR, UK
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, George Mason University, Manassas, VA 20110, USA
| | - Babak Behnam
- Avicenna Biotech Research, Germantown, MD 20871, USA
| |
Collapse
|
24
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
25
|
Wang C, Xu S, Yang X. Hypoxia-Driven Changes in Tumor Microenvironment: Insights into Exosome-Mediated Cell Interactions. Int J Nanomedicine 2024; 19:8211-8236. [PMID: 39157736 PMCID: PMC11328847 DOI: 10.2147/ijn.s479533] [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: 05/23/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024] Open
Abstract
Hypoxia, as a prominent feature of the tumor microenvironment, has a profound impact on the multicomponent changes within this environment. Under hypoxic conditions, the malignant phenotype of tumor cells, the variety of cell types within the tumor microenvironment, as well as intercellular communication and material exchange, undergo complex alterations. These changes provide significant prospects for exploring the mechanisms of tumor development under different microenvironmental conditions and for devising therapeutic strategies. Exosomes secreted by tumor cells and stromal cells are integral components of the tumor microenvironment, serving as crucial mediators of intercellular communication and material exchange, and have consequently garnered increasing attention from researchers. This review focuses on the mechanisms by which hypoxic conditions promote the release of exosomes by tumor cells and alter their encapsulated contents. It also examines the effects of exosomes derived from tumor cells, immune cells, and other cell types under hypoxic conditions on the tumor microenvironment. Additionally, we summarize current research progress on the potential clinical applications of exosomes under hypoxic conditions and propose future research directions in this field.
Collapse
Affiliation(s)
- Churan Wang
- Dalian Medical University, Dalian, 116000, People’s Republic of China
| | - Shun Xu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, 110002, People’s Republic of China
| | - Xiao Yang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, 110002, People’s Republic of China
| |
Collapse
|
26
|
Wang Z, Wang H, Wang Q, Huang T, Guo C, Ji J, Su M, Xu W, Cao Y, Dong Z. Transcriptome analysis of anaerobic glycolysis effects on Jurkat T cell proliferation. Cent Eur J Immunol 2024; 49:194-202. [PMID: 39381560 PMCID: PMC11457565 DOI: 10.5114/ceji.2024.142116] [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: 03/01/2024] [Accepted: 06/11/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction To explore the effects of anaerobic glycolysis on Jurkat T cell proliferation and clarify the possible mechanism via transcriptomic analysis. Material and methods The monocarboxylate transporter 1 inhibitor AZD3965 was used to target and block the transmembrane transport of lactate, thereby inhibiting anaerobic glycolysis in Jurkat T cells. Then, genes with differential expression between treated and untreated cells were detected by transcriptomic analysis, and constructs were generated. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses as well as protein-protein interaction (PPI) network analysis were performed to explore the potential mechanism. Results Inhibition of anaerobic glycolysis reduced Jurkat T-cell proliferation. RNA sequencing identified 1723 transcripts that were differentially expressed, including 1460 upregulated genes and 263 downregulated genes. GO functional enrichment analysis showed that the differentially expressed genes were mainly involved in the biological processes of response to unfolded protein, response to topologically incorrect protein, and protein folding. KEGG pathway analysis of differentially expressed genes or hub genes from the PPI network analysis revealed enrichment in the estrogen signaling and PI3K-Akt pathways. Conclusions Anaerobic glycolysis contributes to the regulation of Jurkat T-cell proliferation. The underlying mechanism may involve the estrogen signaling pathway or PI3K-Akt signaling pathway as well as protein metabolism.
Collapse
Affiliation(s)
- Ziyu Wang
- The Affiliated Hospital of Qingdao University, China
| | - Hongyang Wang
- The Affiliated Hospital of Qingdao University, China
| | - Qinghai Wang
- The Affiliated Hospital of Qingdao University, China
| | - Tao Huang
- The Affiliated Hospital of Qingdao University, China
| | - Chen Guo
- The Affiliated Hospital of Qingdao University, China
| | - Jianlei Ji
- The Affiliated Hospital of Qingdao University, China
| | - Meijie Su
- The Affiliated Hospital of Qingdao University, China
| | - Weijia Xu
- The Affiliated Hospital of Qingdao University, China
| | - Yanwei Cao
- The Affiliated Hospital of Qingdao University, China
| | - Zhen Dong
- The Affiliated Hospital of Qingdao University, China
| |
Collapse
|
27
|
Phull AR, Arain SQ, Majid A, Fatima H, Ahmed M, Kim SJ. Oxidative stress-mediated epigenetic remodeling, metastatic progression and cell signaling in cancer. ONCOLOGIE 2024; 26:493-507. [DOI: 10.1515/oncologie-2024-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Abstract
Cancer is a serious public health issue and cases are rising at a high rate around the world. Altered production of reactive oxygen species (ROS) causes oxidative stress (OS) which plays a vital role in cancer development by disrupting signaling pathways and genomic integrity in the cellular microenvironment. In this study, we reviewed the regulation of noncoding RNAs, histone modifications, and DNA methylation which OS is involved in. These mechanisms promote cancer growth, metastasis, and resistance to chemotherapeutic agents. There is significant potential to improve patient outcomes through the development of customized medications and interventions that precisely address the role of OS in the onset and progression of cancer. Redox-modulating drugs, antioxidant-based therapies, and measures to restore regular cellular activity and OS-modulated signaling pathways are some examples of these strategies. One other hypothesis rationalizes the cancer-suppressing effect of OS, which acts as a two-edged condition that warns against the use of antioxidants for cancer treatment and management. The present study was executed to review the impact of OS on epigenetic machinery, the evolution of metastatic cancer, and how OS mediates cellular signaling. Along with, insights into the potential of targeting OS-mediated mechanisms for cancer therapy.
Collapse
Affiliation(s)
- Abdul-Rehman Phull
- Department of Biochemistry , 66858 Shah Abdul Latif University , Khairpur , Sindh , Pakistan
| | - Sadia Qamar Arain
- Department of Biochemistry , 66858 Shah Abdul Latif University , Khairpur , Sindh , Pakistan
| | - Abdul Majid
- Department of Biochemistry , 66858 Shah Abdul Latif University , Khairpur , Sindh , Pakistan
| | - Humaira Fatima
- Department of Pharmacy , Quaid-i-Azam University , Islamabad , Pakistan
| | - Madiha Ahmed
- Shifa College of Pharmaceutical Sciences , Shifa Tameer-e-Millat University , Islamabad , Pakistan
| | - Song-Ja Kim
- Department of Biological Sciences, College of Natural Sciences , Kongju National University , Gongju , South Korea
| |
Collapse
|
28
|
Zhang M, Zhong J, Song Z, Xu Q, Chen Y, Zhang Z. Regulatory mechanisms and potential therapeutic targets in precancerous lesions of gastric cancer: A comprehensive review. Biomed Pharmacother 2024; 177:117068. [PMID: 39018877 DOI: 10.1016/j.biopha.2024.117068] [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: 05/10/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024] Open
Abstract
Precancerous lesions of gastric cancer (PLGC) represent a critical pathological stage in the transformation from normal gastric mucosa to gastric cancer (GC). The global incidence of PLGC has been rising over the past few decades, with a trend towards younger onset ages. Increasing evidence suggests that early prevention and treatment of PLGC can effectively reverse the malignant development of gastric mucosal epithelial cells. However, there is currently a lack of effective therapeutic drugs and methods. Recent years have witnessed substantial advancements in PLGC research, with the elucidation of novel regulatory mechanisms offering promising avenues for clinical intervention and drug development. This review aims to delineate potential targets for early prevention and diagnosis of GC while exploring innovative approaches to PLGC management. This article focuses on elucidating the regulatory mechanisms of the inflammatory microenvironment, bile acids (BA), glycolysis, autophagy, apoptosis, ferroptosis, and cellular senescence. We pay particular attention to potential therapeutic targets for PLGC, with the goal of providing insights and theoretical basis for clinical research on PLGC.
Collapse
Affiliation(s)
- Maofu Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Jialin Zhong
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhongyang Song
- Department of Oncology, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730020, China
| | - Qian Xu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yuchan Chen
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhiming Zhang
- Department of Oncology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu 730050, China.
| |
Collapse
|
29
|
Zamer BA, Cui ZG, Eladl MA, Hamad M, Muhammad JS. Estrogen treatment in combination with pyruvate kinase M2 inhibition precipitate significant cumulative antitumor effects in colorectal cancer. J Biochem Mol Toxicol 2024; 38:e23799. [PMID: 39132768 DOI: 10.1002/jbt.23799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024]
Abstract
It is well established that pyruvate kinase M2 (PKM2) activity contributes to metabolic reprogramming in various cancers, including colorectal cancer (CRC). Estrogen or 17β-estradiol (E2) signaling is also known to modulate glycolysis markers in cancer cells. However, whether the inhibition of PKM2 combined with E2 treatment could adversely affect glucose metabolism in CRC cells remains to be investigated. First, we confirmed the metabolic plasticity of CRC cells under varying environmental conditions. Next, we identified glycolysis markers that were upregulated in CRC patients and assessed in vitro mRNA levels following E2 treatment. We found that PKM2 expression, which is highly upregulated in CRC clinical samples, is not altered by E2 treatment in CRC cells. In this study, glucose uptake, generation of reactive oxygen species (ROS), lactate production, cell viability, and apoptosis were evaluated in CRC cells following E2 treatment, PKM2 silencing, or a combination of both. Compared to individual treatments, combination therapy resulted in a significant reduction in cell viability and enhanced apoptosis. Glucose uptake and ROS production were markedly reduced in PKM2-silenced E2-treated cells. The data presented here suggest that E2 signaling combined with PKM2 inhibition cumulatively targets glucose metabolism in a manner that negatively impacts CRC cell growth. These findings hold promise for novel therapeutic strategies targeting altered metabolic pathways in CRC.
Collapse
Affiliation(s)
- Batoul Abi Zamer
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research, Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Zheng-Guo Cui
- Department of Environmental Health, School of Medical Sciences, University of Fukui, Fukui, Japan
| | - Mohamed Ahmed Eladl
- Research, Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mawieh Hamad
- Research, Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research, Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biomedical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
30
|
Penarete-Acosta D, Stading R, Emerson L, Horn M, Chakraborty S, Han A, Jayaraman A. A microfluidic co-culture model for investigating colonocytes-microbiota interactions in colorectal cancer. LAB ON A CHIP 2024; 24:3690-3703. [PMID: 38973701 DOI: 10.1039/d4lc00013g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Changes in the abundance of certain bacterial species within the colorectal microbiota correlate with colorectal cancer (CRC) development. While carcinogenic mechanisms of single pathogenic bacteria have been characterized in vitro, limited tools are available to investigate interactions between pathogenic bacteria and both commensal microbiota and colonocytes in a physiologically relevant tumor microenvironment. To address this, we developed a microfluidic device that can be used to co-culture colonocyte spheroids and colorectal microbiota. The device was used to explore the effect of Fusobacterium nucleatum, an opportunistic pathogen associated with colorectal cancer development in humans, on colonocyte gene expression and microbiota composition. F. nucleatum altered the transcription of genes involved in cytokine production, epithelial-to-mesenchymal transition, and proliferation in colonocytes in a contact-independent manner; however, most of these effects were significantly diminished by the presence of commensal microbiota. Interestingly, F. nucleatum significantly altered the abundance of multiple bacterial clades associated with mucosal immune responses and cancer development in the colon. Our results highlight the importance of evaluating the potential carcinogenic activity of pathogens in the context of a commensal microbiota, and the potential to discover novel inter-species microbial interactions in the CRC microenvironment.
Collapse
Affiliation(s)
| | - Rachel Stading
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, USA
| | - Laura Emerson
- Department of Biomedical Engineering, Texas A&M University, USA.
| | - Mitchell Horn
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, College of Medicine, Texas A&M University, USA
| | - Arum Han
- Department of Biomedical Engineering, Texas A&M University, USA.
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, USA
- Department of Electrical and Computer Engineering, Texas A&M University, USA
| | - Arul Jayaraman
- Department of Biomedical Engineering, Texas A&M University, USA.
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, USA
| |
Collapse
|
31
|
Masarkar N, Pal M, Roy M, Yadav AK, Pandya B, Lokhande S, Kanwar JR, Ray SK, Mukherjee S. In-silico screening of bioactive compounds of Moringa oleifera as potential inhibitors targeting HIF-1α/VEGF/GLUT-1 pathway against Breast Cancer. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2024; 0:jcim-2024-0176. [PMID: 39024644 DOI: 10.1515/jcim-2024-0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES Breast cancer is among the most heterogeneous and aggressive diseases and a foremost cause of death in women globally. Hypoxic activation of HIF-1α in breast cancers triggers the transcription of a battery of genes encoding proteins that facilitate tumor growth and metastasis and is correlated with a poor prognosis. Based on the reported cytotoxic and anti-cancer properties of Moringa oleifera (Mo), this study explores the inhibitory effect of bioactive compounds from M. oleifera and breast cancer target proteins HIF-1α, VEGF, and GLUT-1 in silico. METHODS The X-ray crystallographic structures of HIF-1α, VEGF, and GLUT1 were sourced from the Protein Data Bank (PDB) and docked with 70 3D PubChem structures of bioactive compounds of M. oleifera using AutoDock Vina, and binding modes were analyzed using Discovery Studio. Five compounds with the highest binding energies were selected and further drug-likeness, oral bioavailability, ADME, and toxicity profiles were analyzed using SwissADME, ADMETSaR, and ADMETlab 3.0 web server. RESULTS Out of the screened 70 bioactive compounds, the top five compounds with the best binding energies were identified namely Apigenin, Ellagic Acid, Isorhamnetin, Luteolin, and Myricetin with each receptor. Molecular docking results indicated that the ligands interact strongly with the target HIF-1α, VEGF, and GLUT-1 receptors through hydrogen bonds and hydrophobic interactions. These compounds showed favorable drug-like and pharmacokinetic properties, possessed no substantial toxicity, and were fairly bioavailable. CONCLUSIONS Results suggested that the compounds possess strong potential in developing putative lead compounds targeting HIF-1α that are safe natural plant-based drugs against breast cancer.
Collapse
Affiliation(s)
- Neha Masarkar
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| | - Maynak Pal
- Department of Chemistry, National Institute of Technology Manipur, Imphal, India
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology Manipur, Imphal, India
| | - Ashish K Yadav
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| | - Bharati Pandya
- Department of General Surgery, All India Institute of Medical Sciences (AIIMS) Bhopal, Bhopal, India
| | - Suryabhan Lokhande
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| | - Jagat R Kanwar
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| | | | - Sukhes Mukherjee
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| |
Collapse
|
32
|
Januškevičienė I, Petrikaitė V. Exploring doxorubicin transport in 2D and 3D models of MDA-MB-231 sublines: impact of hypoxia and cellular heterogeneity on doxorubicin accumulation in cells. Am J Cancer Res 2024; 14:3584-3599. [PMID: 39113879 PMCID: PMC11301288 DOI: 10.62347/vnwh9165] [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: 05/31/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Triple-negative breast cancer (TNBC) treatment is challenging due to its aggressive nature and heterogeneity of this type of cancer, characterized by various subtypes and intratumoral diversity. Doxorubicin (DOX) plays a crucial role in TNBC chemotherapy reducing the tumor size and improving patient survival. However, decreased drug uptake and increased resistance in specific cell subpopulations reduce the effectiveness of the treatment. This study explored the differences in DOX transport in MDA-MB-231 phenotypic sublines in cell monolayer (2D model) and cell spheroids (3D cultures). Cell spheroids were formed using magnetic 3D Bioprinting method. DOX transport into cells and spheroids was evaluated using fluorescence microscopy after different incubation durations with DOX in normoxia and hypoxia. In hypoxia, DOX transport into cells was 2.5 to 5-fold lower than in normoxia. The subline F5 monolayer-cultured cells exhibited the highest DOX uptake, while subline H2 cells showed the lowest uptake in normoxia and hypoxia. In 3D cultures, DOX transport was up to 2-fold lower in spheroids formed from subline H2 cells. Spheroids from subline D8 and MDA-MB-231 parent cells had the highest DOX uptake. A correlation was observed between the characteristics of the cells and their resistance to anticancer drugs. The results indicate that different cancer cell subpopulations in tumours due to differences in drug uptake could significantly impact treatment efficacy.
Collapse
Affiliation(s)
- Indrė Januškevičienė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Sukilėlių av., LT-50162 Kaunas, Lithuania
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Sukilėlių av., LT-50162 Kaunas, Lithuania
| |
Collapse
|
33
|
Basheeruddin M, Qausain S. Hypoxia-Inducible Factor 1-Alpha (HIF-1α): An Essential Regulator in Cellular Metabolic Control. Cureus 2024; 16:e63852. [PMID: 39099978 PMCID: PMC11297807 DOI: 10.7759/cureus.63852] [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: 06/20/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
The element that causes hypoxia when the von Hippel-Lindau (VHL) protein is not functioning is hypoxia-inducible factor 1-alpha (HIF-1α), which is the essential protein linked to cell control under hypoxia. Consequently, in situations where cells are oxygen-deficient, HIF-1α carries out a variety of essential functions. Citations to relevant literature support the notion that HIF-1α regulates the mitochondrial and glycolytic pathways, as well as the transition from the former to the latter. Cells with limited oxygen supply benefit from this change, which is especially beneficial for the inhibition of the mitochondrial electron transport chain and enhanced uptake of glucose and lactate. During hypoxic stress, HIF-1α also controls proline and glycolytic transporters such as lactate dehydrogenase A (LDHA) and glucose transporter 1 (GLUT1). These mechanisms help the cell return to homeostasis. Therefore, through metabolic change promoting adenosine triphosphate (ATP) synthesis and reducing reactive oxygen species (ROS) creation, HIF-1α may have a role in reducing oxidative stress in cells. This evidence, which describes the function of HIF-1α in many molecular pathways, further supports the notion that it is prognostic and that it contributes to hypoxic cell adaption. Understanding more about disorders, including inflammation, cancer, and ischemia, is possible because of HIF-1α's effect on metabolic changes. Gaining knowledge about the battle between metabolism, which is directed by HIF-1α, would help advance the research on pathophysiological situations involving dysregulated hypoxia and metabolism.
Collapse
Affiliation(s)
- Mohd Basheeruddin
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sana Qausain
- Biomedical Sciences, Allied Health Sciences, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
34
|
Khameneh SC, Razi S, Lashanizadegan R, Akbari S, Sayaf M, Haghani K, Bakhtiyari S. MicroRNA-mediated metabolic regulation of immune cells in cancer: an updated review. Front Immunol 2024; 15:1424909. [PMID: 39007129 PMCID: PMC11239499 DOI: 10.3389/fimmu.2024.1424909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024] Open
Abstract
The study of immunometabolism, which examines how immune cells regulate their metabolism to maintain optimal performance, has become an important area of focus in cancer immunology. Recent advancements in this field have highlighted the intricate connection between metabolism and immune cell function, emphasizing the need for further research. MicroRNAs (miRNAs) have gained attention for their ability to post-transcriptionally regulate gene expression and impact various biological processes, including immune function and cancer progression. While the role of miRNAs in immunometabolism is still being explored, recent studies have demonstrated their significant influence on the metabolic activity of immune cells, such as macrophages, T cells, B cells, and dendritic cells, particularly in cancer contexts. Disrupted immune cell metabolism is a hallmark of cancer progression, and miRNAs have been linked to this process. Understanding the precise impact of miRNAs on immune cell metabolism in cancer is essential for the development of immunotherapeutic approaches. Targeting miRNAs may hold potential for creating groundbreaking cancer immunotherapies to reshape the tumor environment and improve treatment outcomes. In summary, the recognition of miRNAs as key regulators of immune cell metabolism across various cancers offers promising potential for refining cancer immunotherapies. Further investigation into how miRNAs affect immune cell metabolism could identify novel therapeutic targets and lead to the development of innovative cancer immunotherapies.
Collapse
Affiliation(s)
| | - Sara Razi
- Vira Ideators of Modern Science, Tehran, Iran
- Vira Pioneers of Modern Science (VIPOMS), Tehran, Iran
| | | | | | - Masoud Sayaf
- Department of Cellular and Molecular Biology, Faculty of Basic Sciences, Azad University Central Tehran Branch, Tehran, Iran
| | - Karimeh Haghani
- Department of Clinical Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Salar Bakhtiyari
- Department of Clinical Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, United States
| |
Collapse
|
35
|
Arizmendi-Izazaga A, Navarro-Tito N, Jiménez-Wences H, Evaristo-Priego A, Priego-Hernández VD, Dircio-Maldonado R, Zacapala-Gómez AE, Mendoza-Catalán MÁ, Illades-Aguiar B, De Nova Ocampo MA, Salmerón-Bárcenas EG, Leyva-Vázquez MA, Ortiz-Ortiz J. Bioinformatics Analysis Reveals E6 and E7 of HPV 16 Regulate Metabolic Reprogramming in Cervical Cancer, Head and Neck Cancer, and Colorectal Cancer through the PHD2-VHL-CUL2-ELOC-HIF-1α Axis. Curr Issues Mol Biol 2024; 46:6199-6222. [PMID: 38921041 PMCID: PMC11202971 DOI: 10.3390/cimb46060370] [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: 05/02/2024] [Revised: 06/06/2024] [Accepted: 06/15/2024] [Indexed: 06/27/2024] Open
Abstract
Human papillomavirus 16 (HPV 16) infection is associated with several types of cancer, such as head and neck, cervical, anal, and penile cancer. Its oncogenic potential is due to the ability of the E6 and E7 oncoproteins to promote alterations associated with cell transformation. HPV 16 E6 and E7 oncoproteins increase metabolic reprogramming, one of the hallmarks of cancer, by increasing the stability of hypoxia-induced factor 1 α (HIF-1α) and consequently increasing the expression levels of their target genes. In this report, by bioinformatic analysis, we show the possible effect of HPV 16 oncoproteins E6 and E7 on metabolic reprogramming in cancer through the E6-E7-PHD2-VHL-CUL2-ELOC-HIF-1α axis. We proposed that E6 and E7 interact with VHL, CUL2, and ELOC in forming the E3 ubiquitin ligase complex that ubiquitinates HIF-1α for degradation via the proteasome. Based on the information found in the databases, it is proposed that E6 interacts with VHL by blocking its interaction with HIF-1α. On the other hand, E7 interacts with CUL2 and ELOC, preventing their binding to VHL and RBX1, respectively. Consequently, HIF-1α is stabilized and binds with HIF-1β to form the active HIF1 complex that binds to hypoxia response elements (HREs), allowing the expression of genes related to energy metabolism. In addition, we suggest an effect of E6 and E7 at the level of PHD2, VHL, CUL2, and ELOC gene expression. Here, we propose some miRNAs targeting PHD2, VHL, CUL2, and ELOC mRNAs. The effect of E6 and E7 may be the non-hydroxylation and non-ubiquitination of HIF-1α, which may regulate metabolic processes involved in metabolic reprogramming in cancer upon stabilization, non-degradation, and translocation to the nucleus.
Collapse
Affiliation(s)
- Adán Arizmendi-Izazaga
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Hilda Jiménez-Wences
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Adilene Evaristo-Priego
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Víctor Daniel Priego-Hernández
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Roberto Dircio-Maldonado
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Ana Elvira Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Miguel Ángel Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Mónica Ascención De Nova Ocampo
- Escuela Nacional de Medicina y Homeopatía, Programa Institucional de Biomedicina Molecular, Instituto Politécnico Nacional, Guillermo Massieu Helguera No. 239 Col. Fracc. La Escalera-Ticomán, Ciudad de Mexico C.P. 07320, Mexico;
| | - Eric Genaro Salmerón-Bárcenas
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México C.P. 07360, Mexico;
| | - Marco Antonio Leyva-Vázquez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Julio Ortiz-Ortiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| |
Collapse
|
36
|
Ahuja S, Sureka N, Zaheer S. Unraveling the intricacies of cancer-associated fibroblasts: a comprehensive review on metabolic reprogramming and tumor microenvironment crosstalk. APMIS 2024. [PMID: 38873945 DOI: 10.1111/apm.13447] [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: 02/02/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are crucial component of tumor microenvironment (TME) which undergo significant phenotypic changes and metabolic reprogramming, profoundly impacting tumor growth. This review delves into CAF plasticity, diverse origins, and the molecular mechanisms driving their continuous activation. Emphasis is placed on the intricate bidirectional crosstalk between CAFs and tumor cells, promoting cancer cell survival, proliferation, invasion, and immune evasion. Metabolic reprogramming, a cancer hallmark, extends beyond cancer cells to CAFs, contributing to the complex metabolic interplay within the TME. The 'reverse Warburg effect' in CAFs mirrors the Warburg effect, involving the export of high-energy substrates to fuel cancer cells, supporting their rapid proliferation. Molecular regulations by key players like p53, Myc, and K-RAS orchestrate this metabolic adaptation. Understanding the metabolic symbiosis between CAFs and tumor cells opens avenues for targeted therapeutic strategies to disrupt this dynamic crosstalk. Unraveling CAF-mediated metabolic reprogramming provides valuable insights for developing novel anticancer therapies. This comprehensive review consolidates current knowledge, shedding light on CAFs' multifaceted roles in the TME and offering potential targets for future therapies.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
37
|
Kumar P, Lacroix M, Dupré P, Arslan J, Fenou L, Orsetti B, Le Cam L, Racoceanu D, Radulescu O. Deciphering oxygen distribution and hypoxia profiles in the tumor microenvironment: a data-driven mechanistic modeling approach. Phys Med Biol 2024; 69:125023. [PMID: 38815610 DOI: 10.1088/1361-6560/ad524a] [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: 03/08/2024] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
Objective. The distribution of hypoxia within tissues plays a critical role in tumor diagnosis and prognosis. Recognizing the significance of tumor oxygenation and hypoxia gradients, we introduce mathematical frameworks grounded in mechanistic modeling approaches for their quantitative assessment within a tumor microenvironment. By utilizing known blood vasculature, we aim to predict hypoxia levels across different tumor types.Approach. Our approach offers a computational method to measure and predict hypoxia using known blood vasculature. By formulating a reaction-diffusion model for oxygen distribution, we derive the corresponding hypoxia profile.Main results. The framework successfully replicates observed inter- and intra-tumor heterogeneity in experimentally obtained hypoxia profiles across various tumor types (breast, ovarian, pancreatic). Additionally, we propose a data-driven method to deduce partial differential equation models with spatially dependent parameters, which allows us to comprehend the variability of hypoxia profiles within tissues. The versatility of our framework lies in capturing diverse and dynamic behaviors of tumor oxygenation, as well as categorizing states of vascularization based on the dynamics of oxygen molecules, as identified by the model parameters.Significance. The proposed data-informed mechanistic method quantitatively assesses hypoxia in the tumor microenvironment by integrating diverse histopathological data and making predictions across different types of data. The framework provides valuable insights from both modeling and biological perspectives, advancing our comprehension of spatio-temporal dynamics of tumor oxygenation.
Collapse
Affiliation(s)
- P Kumar
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS, INSERM, Montpellier, France
- Sorbonne Université, CNRS, INSERM, AP-HP, Inria, Paris Brain Institute (ICM), Paris, France
| | - M Lacroix
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - P Dupré
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - J Arslan
- Sorbonne Université, CNRS, INSERM, AP-HP, Inria, Paris Brain Institute (ICM), Paris, France
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye & Ear Hospital, East Melbourne, Australia
| | - L Fenou
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - B Orsetti
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - L Le Cam
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - D Racoceanu
- Sorbonne Université, CNRS, INSERM, AP-HP, Inria, Paris Brain Institute (ICM), Paris, France
| | - O Radulescu
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
38
|
Garcia AK, Almodovar S. The Intersection of HIV and Pulmonary Vascular Health: From HIV Evolution to Vascular Cell Types to Disease Mechanisms. JOURNAL OF VASCULAR DISEASES 2024; 3:174-200. [PMID: 39464800 PMCID: PMC11507615 DOI: 10.3390/jvd3020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
People living with HIV (PLWH) face a growing burden of chronic diseases, owing to the combinations of aging, environmental triggers, lifestyle choices, and virus-induced chronic inflammation. The rising incidence of pulmonary vascular diseases represents a major concern for PLWH. The study of HIV-associated pulmonary vascular complications ideally requires a strong understanding of pulmonary vascular cell biology and HIV pathogenesis at the molecular level for effective applications in infectious diseases and vascular medicine. Active HIV infection and/or HIV proteins disturb the delicate balance between vascular tone and constriction, which is pivotal for maintaining pulmonary vascular health. One of the defining features of HIV is its high genetic diversity owing to several factors including its high mutation rate, recombination between viral strains, immune selective pressures, or even geographical factors. The intrinsic HIV genetic diversity has several important implications for pathogenic outcomes of infection and the overall battle to combat HIV. Challenges in the field present themselves from two sides of the same coin: those imposed by the virus itself and those stemming from the host. The field may be advanced by further developing in vivo and in vitro models that are well described for both pulmonary vascular diseases and HIV for mechanistic studies. In essence, the study of HIV-associated pulmonary vascular complications requires a multidisciplinary approach, drawing upon insights from both infectious diseases and vascular medicine. In this review article, we discuss the fundamentals of HIV virology and their impact on pulmonary disease, aiming to enhance the understanding of either area or both simultaneously. Bridging the gap between preclinical research findings and clinical practice is essential for improving patient care. Addressing these knowledge gaps requires interdisciplinary collaborations, innovative research approaches, and dedicated efforts to prioritize HIV-related pulmonary complications on the global research agenda.
Collapse
Affiliation(s)
- Amanda K. Garcia
- Department of Immunology & Molecular Microbiology, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX 79430, USA
| | - Sharilyn Almodovar
- Department of Immunology & Molecular Microbiology, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX 79430, USA
- Center for Tropical Medicine & Infectious Diseases, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX 79430, USA
| |
Collapse
|
39
|
Nam A, Jain S, Wu C, Campos A, Shepard RM, Yu Z, Reddy JP, Von Schalscha T, Weis SM, Onaitis M, Wettersten HI, Cheresh DA. Integrin αvβ3 Upregulation in Response to Nutrient Stress Promotes Lung Cancer Cell Metabolic Plasticity. Cancer Res 2024; 84:1630-1642. [PMID: 38588407 PMCID: PMC11096068 DOI: 10.1158/0008-5472.can-23-2700] [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: 09/05/2023] [Revised: 01/05/2024] [Accepted: 02/16/2024] [Indexed: 04/10/2024]
Abstract
Cancer stem/tumor-initiating cells display stress tolerance and metabolic flexibility to survive in a harsh environment with limited nutrient and oxygen availability. The molecular mechanisms underlying this phenomenon could provide targets to prevent metabolic adaptation and halt cancer progression. Here, we showed in cultured cells and live human surgical biopsies of non-small cell lung cancer that nutrient stress drives the expression of the epithelial cancer stem cell marker integrin αvβ3 via upregulation of the β3 subunit, resulting in a metabolic reprogramming cascade that allows tumor cells to thrive despite a nutrient-limiting environment. Although nutrient deprivation is known to promote acute, yet transient, activation of the stress sensor AMP-activated protein kinase (AMPK), stress-induced αvβ3 expression via Src activation unexpectedly led to secondary and sustained AMPK activation. This resulted in the nuclear localization of peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC1α) and upregulation of glutamine metabolism, the tricarboxylic acid cycle, and oxidative phosphorylation. Pharmacological or genetic targeting of this axis prevented lung cancer cells from evading the effects of nutrient stress, thereby blocking tumor initiation in mice following orthotopic implantation of lung cancer cells. These findings reveal a molecular pathway driven by nutrient stress that results in cancer stem cell reprogramming to promote metabolic flexibility and tumor initiation. SIGNIFICANCE Upregulation of integrin αvβ3, a cancer stem cell marker, in response to nutrient stress activates sustained AMPK/PGC1α signaling that induces metabolic reprogramming in lung cancer cells to support their survival. See related commentary by Rainero, p. 1543.
Collapse
Affiliation(s)
- Arin Nam
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - Shashi Jain
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
- Department of Neurology, University of California, Irvine, Irvine, California, United States of America
| | - Chengsheng Wu
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - Alejandro Campos
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - Ryan M. Shepard
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - Ziqi Yu
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - Joshua P. Reddy
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - Tami Von Schalscha
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - Sara M. Weis
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - Mark Onaitis
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - Hiromi I. Wettersten
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
| | - David A. Cheresh
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, California, United States of America
- Lead contact
| |
Collapse
|
40
|
Beauchamp E, Gamma JM, Cromwell CR, Moussa EW, Pain R, Kostiuk MA, Acevedo-Morantes C, Iyer A, Yap M, Vincent KM, Postovit LM, Julien O, Hubbard BP, Mackey JR, Berthiaume LG. Multiomics analysis identifies oxidative phosphorylation as a cancer vulnerability arising from myristoylation inhibition. J Transl Med 2024; 22:431. [PMID: 38715059 PMCID: PMC11075276 DOI: 10.1186/s12967-024-05150-6] [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] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/31/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND In humans, two ubiquitously expressed N-myristoyltransferases, NMT1 and NMT2, catalyze myristate transfer to proteins to facilitate membrane targeting and signaling. We investigated the expression of NMTs in numerous cancers and found that NMT2 levels are dysregulated by epigenetic suppression, particularly so in hematologic malignancies. This suggests that pharmacological inhibition of the remaining NMT1 could allow for the selective killing of these cells, sparing normal cells with both NMTs. METHODS AND RESULTS Transcriptomic analysis of 1200 NMT inhibitor (NMTI)-treated cancer cell lines revealed that NMTI sensitivity relates not only to NMT2 loss or NMT1 dependency, but also correlates with a myristoylation inhibition sensitivity signature comprising 54 genes (MISS-54) enriched in hematologic cancers as well as testis, brain, lung, ovary, and colon cancers. Because non-myristoylated proteins are degraded by a glycine-specific N-degron, differential proteomics revealed the major impact of abrogating NMT1 genetically using CRISPR/Cas9 in cancer cells was surprisingly to reduce mitochondrial respiratory complex I proteins rather than cell signaling proteins, some of which were also reduced, albeit to a lesser extent. Cancer cell treatments with the first-in-class NMTI PCLX-001 (zelenirstat), which is undergoing human phase 1/2a trials in advanced lymphoma and solid tumors, recapitulated these effects. The most downregulated myristoylated mitochondrial protein was NDUFAF4, a complex I assembly factor. Knockout of NDUFAF4 or in vitro cell treatment with zelenirstat resulted in loss of complex I, oxidative phosphorylation and respiration, which impacted metabolomes. CONCLUSIONS Targeting of both, oxidative phosphorylation and cell signaling partly explains the lethal effects of zelenirstat in select cancer types. While the prognostic value of the sensitivity score MISS-54 remains to be validated in patients, our findings continue to warrant the clinical development of zelenirstat as cancer treatment.
Collapse
Affiliation(s)
| | - Jay M Gamma
- Department of Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Christopher R Cromwell
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Eman W Moussa
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rony Pain
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Morris A Kostiuk
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Claudia Acevedo-Morantes
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Aishwarya Iyer
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Megan Yap
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Krista M Vincent
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Lynne M Postovit
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Olivier Julien
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Basil P Hubbard
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | | | - Luc G Berthiaume
- Pacylex Pharmaceuticals Inc., Edmonton, AB, Canada.
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
41
|
Chowdhury M, Das PK. Hypoxia: Intriguing Feature in Cancer Cell Biology. ChemMedChem 2024; 19:e202300551. [PMID: 38328976 DOI: 10.1002/cmdc.202300551] [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: 10/14/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 02/09/2024]
Abstract
Hypoxia, a key aspect of the tumor microenvironment, plays a vital role in cell proliferation, angiogenesis, metabolism, and the immune response within tumors. These factors collectively promote tumor advancement, aggressiveness, metastasis and result in a poor prognosis. Hypoxia inducible factor 1α (HIF-1α), activated under low oxygen conditions, mediates many of these effects by altering drug target expression, metabolic regulation, and oxygen consumption. These changes promote cancer cell growth and survival. Hypoxic tumor cells develop aggressive traits and resistance to chemotherapy and radiotherapy, leading to increased mortality. Targeting hypoxic tumor offers a potential solution to overcome the challenges posed by tumor heterogeneity and can be used in designing diagnostic and therapeutic nanocarriers for various solid cancers. This concept provides an overview of the intricate relationship between hypoxia and the tumor microenvironment, highlighting its potential as a promising tool for cancer therapies. The article explores the development of hypoxia in cancer cells and its role in cancer progression, along with the latest advancements in hypoxia-triggered cancer treatment.
Collapse
Affiliation(s)
- Monalisa Chowdhury
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700 032, India
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700 032, India
| |
Collapse
|
42
|
Chinopoulos C. Complex I activity in hypoxia: implications for oncometabolism. Biochem Soc Trans 2024; 52:529-538. [PMID: 38526218 PMCID: PMC11088919 DOI: 10.1042/bst20230189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
Certain cancer cells within solid tumors experience hypoxia, rendering them incapable of oxidative phosphorylation (OXPHOS). Despite this oxygen deficiency, these cells exhibit biochemical pathway activity that relies on NAD+. This mini-review scrutinizes the persistent, residual Complex I activity that oxidizes NADH in the absence of oxygen as the electron acceptor. The resulting NAD+ assumes a pivotal role in fueling the α-ketoglutarate dehydrogenase complex, a critical component in the oxidative decarboxylation branch of glutaminolysis - a hallmark oncometabolic pathway. The proposition is that through glutamine catabolism, high-energy phosphate intermediates are produced via substrate-level phosphorylation in the mitochondrial matrix substantiated by succinyl-CoA ligase, partially compensating for an OXPHOS deficiency. These insights provide a rationale for exploring Complex I inhibitors in cancer treatment, even when OXPHOS functionality is already compromised.
Collapse
|
43
|
Weng X, Ma T, Chen Q, Chen BW, Shan J, Chen W, Zhi X. Decreased expression of H19/miR-675 ameliorates hypoxia-induced oxaliplatin resistance in colorectal cancer. Heliyon 2024; 10:e27027. [PMID: 38449593 PMCID: PMC10915565 DOI: 10.1016/j.heliyon.2024.e27027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/25/2024] [Accepted: 02/22/2024] [Indexed: 03/08/2024] Open
Abstract
Hypoxic microenvironment, a hallmark of solid tumors, contributes to chemoresistance, and long noncoding (lnc) RNAs are involved in hypoxia-induced drug resistance. However, the role of lncRNAs in hypoxic tumor chemotherapy resistance remains unclear. Here, we aimed to elucidate the effects of lncRNAs in hypoxia-mediated resistance in colorectal cancer (CRC), as well as the underlying mechanisms. The results indicated that the expression of lncRNA H19 was enhanced in hypoxia- or oxaliplatin-treated CRC cells; moreover, H19 contributed to drug resistance in CRC cells both in vitro and in vivo. Mechanistically, H19 was noted to act as a competitive endogenous RNA of miR-675-3p to regulate epithelial-mesenchymal transition (EMT). Notably, an miR-675-3p mimic could attenuate the effects of H19 deficiency in CRC cells with hypoxia-induced chemoresistance. In conclusion, H19 downregulation may counteract hypoxia-induced chemoresistance by sponging miR-675-3p to regulate EMT; as such, the H19/miR-675-3p axis might be a promising therapeutic target for drug resistance in CRC.
Collapse
Affiliation(s)
- Xingyue Weng
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Tao Ma
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Qi Chen
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Bryan Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Jianzhen Shan
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang Province, China
| | - Xiao Zhi
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, Zhejiang, 310003, China
| |
Collapse
|
44
|
Simula L, Fumagalli M, Vimeux L, Rajnpreht I, Icard P, Birsen G, An D, Pendino F, Rouault A, Bercovici N, Damotte D, Lupo-Mansuet A, Alifano M, Alves-Guerra MC, Donnadieu E. Mitochondrial metabolism sustains CD8 + T cell migration for an efficient infiltration into solid tumors. Nat Commun 2024; 15:2203. [PMID: 38467616 PMCID: PMC10928223 DOI: 10.1038/s41467-024-46377-7] [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: 08/13/2023] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
The ability of CD8+ T cells to infiltrate solid tumors and reach cancer cells is associated with improved patient survival and responses to immunotherapy. Thus, identifying the factors controlling T cell migration in tumors is critical, so that strategies to intervene on these targets can be developed. Although interstitial motility is a highly energy-demanding process, the metabolic requirements of CD8+ T cells migrating in a 3D environment remain unclear. Here, we demonstrate that the tricarboxylic acid (TCA) cycle is the main metabolic pathway sustaining human CD8+ T cell motility in 3D collagen gels and tumor slices while glycolysis plays a more minor role. Using pharmacological and genetic approaches, we report that CD8+ T cell migration depends on the mitochondrial oxidation of glucose and glutamine, but not fatty acids, and both ATP and ROS produced by mitochondria are required for T cells to migrate. Pharmacological interventions to increase mitochondrial activity improve CD8+ T cell intratumoral migration and CAR T cell recruitment into tumor islets leading to better control of tumor growth in human xenograft models. Our study highlights the rationale of targeting mitochondrial metabolism to enhance the migration and antitumor efficacy of CAR T cells in treating solid tumors.
Collapse
Affiliation(s)
- Luca Simula
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Equipe labellisée "Ligue contre le Cancer", Paris, 75014, France.
| | - Mattia Fumagalli
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Equipe labellisée "Ligue contre le Cancer", Paris, 75014, France
| | - Lene Vimeux
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Equipe labellisée "Ligue contre le Cancer", Paris, 75014, France
| | - Irena Rajnpreht
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Equipe labellisée "Ligue contre le Cancer", Paris, 75014, France
| | - Philippe Icard
- Université de Normandie, UNICAEN, Inserm U1086 Interdisciplinary Research Unit for Cancer Prevention and Treatment, Caen, France
- Thoracic Surgery Department, Cochin Hospital, APHP-Centre, Université Paris-Cité, Paris, France
| | - Gary Birsen
- Department of Pneumology, Thoracic Oncology Unit, Cochin Hospital, APHP-Centre, Université Paris-Cité, 75014, Paris, France
| | - Dongjie An
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Equipe labellisée "Ligue contre le Cancer", Paris, 75014, France
| | - Frédéric Pendino
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Equipe labellisée "Ligue contre le Cancer", Paris, 75014, France
| | - Adrien Rouault
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Equipe labellisée "Ligue contre le Cancer", Paris, 75014, France
| | - Nadège Bercovici
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Equipe labellisée "Ligue contre le Cancer", Paris, 75014, France
| | - Diane Damotte
- Department of Pathology, Cochin Hospital, APHP-Centre, Université Paris-Cité, 75014, Paris, France
| | - Audrey Lupo-Mansuet
- Department of Pathology, Cochin Hospital, APHP-Centre, Université Paris-Cité, 75014, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP-Centre, Université Paris-Cité, Paris, France
- Inserm U1138, Integrative Cancer Immunology Unit, 75006, Paris, France
| | | | - Emmanuel Donnadieu
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Equipe labellisée "Ligue contre le Cancer", Paris, 75014, France.
| |
Collapse
|
45
|
Liao M, Yao D, Wu L, Luo C, Wang Z, Zhang J, Liu B. Targeting the Warburg effect: A revisited perspective from molecular mechanisms to traditional and innovative therapeutic strategies in cancer. Acta Pharm Sin B 2024; 14:953-1008. [PMID: 38487001 PMCID: PMC10935242 DOI: 10.1016/j.apsb.2023.12.003] [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: 07/05/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer reprogramming is an important facilitator of cancer development and survival, with tumor cells exhibiting a preference for aerobic glycolysis beyond oxidative phosphorylation, even under sufficient oxygen supply condition. This metabolic alteration, known as the Warburg effect, serves as a significant indicator of malignant tumor transformation. The Warburg effect primarily impacts cancer occurrence by influencing the aerobic glycolysis pathway in cancer cells. Key enzymes involved in this process include glucose transporters (GLUTs), HKs, PFKs, LDHs, and PKM2. Moreover, the expression of transcriptional regulatory factors and proteins, such as FOXM1, p53, NF-κB, HIF1α, and c-Myc, can also influence cancer progression. Furthermore, lncRNAs, miRNAs, and circular RNAs play a vital role in directly regulating the Warburg effect. Additionally, gene mutations, tumor microenvironment remodeling, and immune system interactions are closely associated with the Warburg effect. Notably, the development of drugs targeting the Warburg effect has exhibited promising potential in tumor treatment. This comprehensive review presents novel directions and approaches for the early diagnosis and treatment of cancer patients by conducting in-depth research and summarizing the bright prospects of targeting the Warburg effect in cancer.
Collapse
Affiliation(s)
- Minru Liao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dahong Yao
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
| | - Lifeng Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chaodan Luo
- Department of Psychology, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhiwen Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jin Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
46
|
Rasouli M, Fattahi R, Nuoroozi G, Zarei-Behjani Z, Yaghoobi M, Hajmohammadi Z, Hosseinzadeh S. The role of oxygen tension in cell fate and regenerative medicine: implications of hypoxia/hyperoxia and free radicals. Cell Tissue Bank 2024; 25:195-215. [PMID: 37365484 DOI: 10.1007/s10561-023-10099-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
Oxygen pressure plays an integral role in regulating various aspects of cellular biology. Cell metabolism, proliferation, morphology, senescence, metastasis, and angiogenesis are some instances that are affected by different tensions of oxygen. Hyperoxia or high oxygen concentration, enforces the production of reactive oxygen species (ROS) that disturbs physiological homeostasis, and consequently, in the absence of antioxidants, cells and tissues are directed to an undesired fate. On the other side, hypoxia or low oxygen concentration, impacts cell metabolism and fate strongly through inducing changes in the expression level of specific genes. Thus, understanding the precise mechanism and the extent of the implication of oxygen tension and ROS in biological events is crucial to maintaining the desired cell and tissue function for application in regenerative medicine strategies. Herein, a comprehensive literature review has been performed to find out the impacts of oxygen tensions on the various behaviors of cells or tissues.
Collapse
Affiliation(s)
- Mehdi Rasouli
- Student Research Committee, Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Fattahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Ghader Nuoroozi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Zeinab Zarei-Behjani
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maliheh Yaghoobi
- Engineering Department, Faculty of Chemical Engineering, Zanjan University, Zanjan, Iran
| | - Zeinab Hajmohammadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran.
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
47
|
Pei YX, Su CG, Liao Z, Li WW, Wang ZX, Liu JL. Comparative effectiveness of several adjuvant therapies after hepatectomy for hepatocellular carcinoma patients with microvascular invasion. World J Gastrointest Surg 2024; 16:554-570. [PMID: 38463369 PMCID: PMC10921205 DOI: 10.4240/wjgs.v16.i2.554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/24/2023] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND For resectable hepatocellular carcinoma (HCC), radical hepatectomy is commonly used as a curative treatment. However, postoperative recurrence significantly diminishes the overall survival (OS) of HCC patients, especially with microvascular invasion (MVI) as an independent high-risk factor for recurrence. While some studies suggest that postoperative adjuvant therapy may decrease the risk of recurrence following liver resection in HCC patients, the specific role of adjuvant therapies in those with MVI remains unclear. AIM To conduct a network meta-analysis (NMA) to evaluate the efficacy of various adjuvant therapies and determine the optimal adjuvant regimen. METHODS A systematic literature search was conducted on PubMed, EMBASE, and Web of Science until April 6, 2023. Studies comparing different adjuvant therapies or comparing adjuvant therapy with hepatectomy alone were included. Hazard ratios (HRs) with 95% confidence intervals were used to combine data on recurrence free survival and OS in both pairwise meta-analyses and NMA. RESULTS Fourteen eligible trials (2268 patients) reporting five different therapies were included. In terms of reducing the risk of recurrence, radiotherapy (RT) [HR = 0.34 (0.23, 0.5); surface under the cumulative ranking curve (SUCRA) = 97.7%] was found to be the most effective adjuvant therapy, followed by hepatic artery infusion chemotherapy [HR = 0.52 (0.35, 0.76); SUCRA = 65.1%]. Regarding OS improvement, RT [HR: 0.35 (0.2, 0.61); SUCRA = 93.1%] demonstrated the highest effectiveness, followed by sorafenib [HR = 0.48 (0.32, 0.69); SUCRA = 70.9%]. CONCLUSION Adjuvant therapy following hepatectomy may reduce the risk of recurrence and provide a survival benefit for HCC patients with MVI. RT appears to be the most effective adjuvant regimen.
Collapse
Affiliation(s)
- Yin-Xuan Pei
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei Province, China
| | - Chen-Guang Su
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei Province, China
| | - Zheng Liao
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei Province, China
| | - Wei-Wei Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei Province, China
| | - Zi-Xiang Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei Province, China
| | - Jin-Long Liu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei Province, China
| |
Collapse
|
48
|
Li R, Wen X, Lv RX, Ren XY, Cheng BL, Wang YK, Chen RZ, Hu W, Tang XR. DNA-methylome-derived epigenetic fingerprint as an immunophenotype indicator of durable clinical immunotherapeutic benefits in head and neck squamous cell carcinoma. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00917-x. [PMID: 38315286 DOI: 10.1007/s13402-024-00917-x] [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] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Cancer immunotherapy provides durable response and improves survival in a subset of head and neck squamous cell carcinoma (HNSC) patients, which may due to discriminative tumor microenvironment (TME). Epigenetic regulations play critical roles in HNSC tumorigenesis, progression, and activation of functional immune cells. This study aims to identify an epigenetic signature as an immunophenotype indicator of durable clinical immunotherapeutic benefits in HNSC patients. METHODS Unsupervised consensus clustering approach was applied to distinguish immunophenotypes based on five immune signatures in The Cancer Genome Atlas (TCGA) HNSC cohort. Two immunophenotypes (immune 'Hot' and immune 'Cold') that had different TME features, diverse prognosis, and distinct DNA methylation patterns were recognized. Immunophenotype-related methylated signatures (IPMS) were identified by the least absolute shrinkage and selector operation algorithm. Additionally, the IPMS score by deconvolution algorithm was constructed as an immunophenotype classifier to predict clinical outcomes and immunotherapeutic response. RESULTS The 'Hot' HNSC immunophenotype had higher immunoactivity and better overall survival (p = 0.00055) compared to the 'Cold' tumors. The immunophenotypes had distinct DNA methylation patterns, which was closely associated with HNSC tumorigenesis and functional immune cell infiltration. 311 immunophenotype-related methylated CpG sites (IRMCs) was identified from TCGA-HNSC dataset. IPMS score model achieved a strong clinical predictive performance for classifying immunophenotypes. The area under the curve value (AUC) of the IPMS score model reached 85.9% and 89.8% in TCGA train and test datasets, respectively, and robustness was verified in five HNSC validation datasets. It was also validated as an immunophenotype classifier for predicting durable clinical benefits (DCB) in lung cancer patients who received anti-PD-1/PD-L1 immunotherapy (p = 0.017) and TCGA-SKCM patients who received distinct immunotherapy (p = 0.033). CONCLUSIONS This study systematically analyzed DNA methylation patterns in distinct immunophenotypes to identify IPMS with clinical prognostic potential for personalized epigenetic anticancer approaches in HNSC patients. The IPMS score model may serve as a reliable epigenome prognostic tool for clinical immunophenotyping to guide immunotherapeutic strategies in HNSC.
Collapse
Affiliation(s)
- Rui Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Xin Wen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Ru-Xue Lv
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Xian-Yue Ren
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Bing-Lin Cheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Yi-Kai Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Ru-Zhen Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Wen Hu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Xin-Ran Tang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China.
| |
Collapse
|
49
|
Yang J, Shay C, Saba NF, Teng Y. Cancer metabolism and carcinogenesis. Exp Hematol Oncol 2024; 13:10. [PMID: 38287402 PMCID: PMC10826200 DOI: 10.1186/s40164-024-00482-x] [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: 10/23/2023] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
Metabolic reprogramming is an emerging hallmark of cancer cells, enabling them to meet increased nutrient and energy demands while withstanding the challenging microenvironment. Cancer cells can switch their metabolic pathways, allowing them to adapt to different microenvironments and therapeutic interventions. This refers to metabolic heterogeneity, in which different cell populations use different metabolic pathways to sustain their survival and proliferation and impact their response to conventional cancer therapies. Thus, targeting cancer metabolic heterogeneity represents an innovative therapeutic avenue with the potential to overcome treatment resistance and improve therapeutic outcomes. This review discusses the metabolic patterns of different cancer cell populations and developmental stages, summarizes the molecular mechanisms involved in the intricate interactions within cancer metabolism, and highlights the clinical potential of targeting metabolic vulnerabilities as a promising therapeutic regimen. We aim to unravel the complex of metabolic characteristics and develop personalized treatment approaches to address distinct metabolic traits, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Jianqiang Yang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Chloe Shay
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
50
|
Mathew M, Nguyen NT, Bhutia YD, Sivaprakasam S, Ganapathy V. Metabolic Signature of Warburg Effect in Cancer: An Effective and Obligatory Interplay between Nutrient Transporters and Catabolic/Anabolic Pathways to Promote Tumor Growth. Cancers (Basel) 2024; 16:504. [PMID: 38339256 PMCID: PMC10854907 DOI: 10.3390/cancers16030504] [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: 12/17/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Aerobic glycolysis in cancer cells, originally observed by Warburg 100 years ago, which involves the production of lactate as the end product of glucose breakdown even in the presence of adequate oxygen, is the foundation for the current interest in the cancer-cell-specific reprograming of metabolic pathways. The renewed interest in cancer cell metabolism has now gone well beyond the original Warburg effect related to glycolysis to other metabolic pathways that include amino acid metabolism, one-carbon metabolism, the pentose phosphate pathway, nucleotide synthesis, antioxidant machinery, etc. Since glucose and amino acids constitute the primary nutrients that fuel the altered metabolic pathways in cancer cells, the transporters that mediate the transfer of these nutrients and their metabolites not only across the plasma membrane but also across the mitochondrial and lysosomal membranes have become an integral component of the expansion of the Warburg effect. In this review, we focus on the interplay between these transporters and metabolic pathways that facilitates metabolic reprogramming, which has become a hallmark of cancer cells. The beneficial outcome of this recent understanding of the unique metabolic signature surrounding the Warburg effect is the identification of novel drug targets for the development of a new generation of therapeutics to treat cancer.
Collapse
Affiliation(s)
| | | | | | | | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (N.T.N.); (Y.D.B.); (S.S.)
| |
Collapse
|