1
|
Fang H, Yu E, Liu C, Eapen C, Cheng C, Hu T. Metabolic landscape and rewiring in normal hematopoiesis, leukemia and aging. Semin Cancer Biol 2025; 111:1-15. [PMID: 39933639 DOI: 10.1016/j.semcancer.2025.02.003] [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: 10/23/2024] [Revised: 01/06/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Recent advancements in metabolism research have demonstrated its critical roles in a lot of critical biological processes, including stemness maintenance, cell differentiation, proliferation, and function. Hematopoiesis is the fundamental cell differentiation process with the production of millions of red blood cells per second in carrying oxygen and white blood cells in fighting infection and cancers. The differentiation processes of hematopoietic stem and progenitor cells (HSPCs) are accompanied by significant metabolic reprogramming. In hematological malignancy, metabolic reprogramming is also essential to the malignant hematopoiesis processes. The metabolic rewiring is driven by distinct molecular mechanisms that meet the specific demands of different target cells. Leukemic cells, for instance, adopt unique metabolic profiles to support their heightened energy needs for survival and proliferation. Moreover, aging HSPCs exhibit altered energy consumption compared to their younger counterparts, often triggering protective mechanisms at the cellular level. In this review, we provide a comprehensive analysis of the metabolic processes involved in hematopoiesis and the metabolic rewiring that occurs under adverse conditions. In addition, we highlight current research directions and discuss the potential of targeting metabolic pathways for the management of hematological malignancies and aging.
Collapse
Affiliation(s)
- Hui Fang
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States; Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Enze Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa 999078, Macao
| | - Chang Liu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States; Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Christy Eapen
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States
| | - Chunming Cheng
- Stephenson Cancer Center at Oklahoma University, Oklahoma City, OK 73104, United States.
| | - Tianxiang Hu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States.
| |
Collapse
|
2
|
Allen TP, Roennfeldt AE, Reckdharajkumar M, Sullivan AE, Liu M, Quinn RJ, Russell DL, Peet DJ, Whitelaw ML, Bersten DC. dFLASH; dual FLuorescent transcription factor activity sensor for histone integrated live-cell reporting and high-content screening. Nat Commun 2025; 16:3298. [PMID: 40195317 PMCID: PMC11977238 DOI: 10.1038/s41467-025-58488-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: 12/09/2023] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Live-cell transcription factor (TF) activity reporting is crucial for synthetic biology, drug discovery and functional genomics. Here we present dFLASH (dual FLuorescent transcription factor Activity Sensor for Histone-integrated live-cell reporting), a modular, genome-integrated TF sensor. dFLASH homogeneously and specifically detects endogenous Hypoxia Inducible Factor (HIF) and Progesterone Receptor (PGR) activities, as well as coactivator recruitment to synthetic TFs. The dFLASH system produces dual-color nuclear fluorescence, enabling normalized, dynamic, live-cell TF activity sensing with strong signal-to-noise ratios and robust screening performance (Z' = 0.61-0.74). We validate dFLASH for functional genomics and drug screening, demonstrating HIF regulation via CRISPRoff and application to whole-genome CRISPR KO screening. Additionally, we apply dFLASH for drug discovery, identifying HIF pathway modulators from a 1600-compound natural product library using high-content imaging. Together, this versatile platform provides a powerful tool for studying TF activity across diverse applications.
Collapse
Affiliation(s)
- Timothy P Allen
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Alison E Roennfeldt
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | | | - Adrienne E Sullivan
- Adelaide Centre for Epigenetics, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- South Australian immunoGENomics Cancer Institute (SAiGENCI), Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Miaomiao Liu
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
| | - Ronald J Quinn
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
| | - Darryl L Russell
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Murray L Whitelaw
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore, 308433, Singapore
| | - David C Bersten
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
3
|
Xiang X, Shao Y, Xiang L, Jiao Q, Zhang W, Qin Y, Chen Y. Suppression of Liver Fibrogenesis with Photothermal Sorafenib Nanovesicles via Selectively Inhibiting Glycolysis and Amplification of Active HSCs. Mol Pharm 2025; 22:1939-1957. [PMID: 40053386 DOI: 10.1021/acs.molpharmaceut.4c01135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2025]
Abstract
As the major driving factor of hepatic fibrosis, the activated hepatic stellate cells (aHSCs) rely on active glycolysis to support their aberrant proliferation and secretion of the extracellular matrix. Sorafenib (Sor) can combat liver fibrosis by suppressing HIF-1α and glycolysis, but its poor solubility, rapid metabolism, and low bioavailability restrict such a clinical application. Here, Sor was loaded onto polydopamine nanoparticles and then encapsulated by a retinoid-decorated red blood cell membrane, yielding HSC-targeted Sor nanovesicles (PDA/Sor@RMV-VA) with a high Sor-loading capacity and photothermally controlled drug release for antifibrotic treatment. These Sor RMVs not only exhibited a good particle size, dispersity and biocompatibility, prolonged circulation time, enhanced aHSC targetability, and hepatic accumulation both in vitro and in vivo, but also displayed a mild photothermal activity proper for promoting sorafenib release and accumulation in CCl4-induced fibrotic mouse livers without incurring phototoxicity. Compared with nontargeting Sor formulations, PDA/Sor@RMV-VA more effectively downregulated HIF-1α and glycolytic enzyme in both cultured aHSCs and fibrotic mice and reversed myofibroblast phenotype and amplification of aHSCs and thus more significantly improved liver damage, inflammation, and fibrosis, all of which could be even further advanced with NIR irradiation. These results fully demonstrate the antifibrotic power and therapeutic potential of PDA/Sor@RMV-VA as an antifibrotic nanomedicine, which would support a new clinical treatment for hepatic fibrosis.
Collapse
Affiliation(s)
- Xianjing Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yaru Shao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Li Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Qiangqiang Jiao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Wenhui Zhang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yuting Qin
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yuping Chen
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
- MOE Key Laboratory of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| |
Collapse
|
4
|
Yang C, Yu R, Zhang Y, Wang Q, Huang D, Cheng Y, Zhu Y, Shen X, Shi Y, Zhao YZ, Yao Q. Curcumin-loaded bioadhesive silk fibroin microsphere improves islet transplantation by mitigating oxidative stress and inhibiting apoptosis. Mater Today Bio 2025; 31:101507. [PMID: 39925714 PMCID: PMC11804778 DOI: 10.1016/j.mtbio.2025.101507] [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: 01/02/2025] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 02/11/2025] Open
Abstract
Islet transplantation, a form of cell therapy involving the injection of healthy islet cells into the recipient's body for curative treatment of T1DM, often fails due to oxidative stress and inflammatory damage experienced by the transplanted islets. Curcumin, a naturally occurring polyphenol with potent anti-inflammatory and antioxidant properties, has been underutilized in islet protection due to challenges associated with its co-delivery and formulation. In this study, we developed bioadhesive curcumin microspheres (PDA/CUR@SF) by incorporating curcumin into a silk fibroin matrix and subsequently coating it with polydopamine to enhance islet adherence. Silk protein acts as a delivery carrier while polydopamine serves as an adhesive agent; both components synergistically provide anti-inflammatory effects. In vitro experiments demonstrated that PDA/CUR@SF enhances islet resistance against oxidative stress and inflammatory damage by improving cell viability and function. In vivo studies showed that PDA/CUR@SF prolongs stabilization of blood glucose levels in diabetic mice receiving islet transplantation while facilitating faster glucose clearance. Further analysis revealed that PDA/CUR@SF protects transplanted islets through co-grafting and promotes neovascularization. Overall, our findings suggest that PDA/CUR@SF offers a rational approach to improve outcomes in transplantation.
Collapse
Affiliation(s)
- Chunhui Yang
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
- The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, 400060, China
| | - Runjie Yu
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ying Zhang
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qian Wang
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| | - Di Huang
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yang Cheng
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yixuan Zhu
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xinyue Shen
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yifan Shi
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qing Yao
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
5
|
Hongyu L, Nan Y, Kaiying L, Zhenning Z, Lili Z, Jing M, Huisheng M. Assessment of Electroacupuncture Therapy with Distant-Approximal Acupoints Based on the HPT Axis in Rats with Oligoasthenospermia Through Transcriptomic Analysis. Reprod Sci 2025; 32:1228-1240. [PMID: 40016484 PMCID: PMC11978543 DOI: 10.1007/s43032-025-01821-x] [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: 12/02/2024] [Accepted: 02/13/2025] [Indexed: 03/01/2025]
Abstract
The transcriptomic analysis was used to explore the effect of electroacupuncture therapy with distant-approximal acupoints based on the hypothalamic-pituitary-testicular (HPT) on gene expression patterns and pathways in oligoasthenospermia (OAT) rats. In this study, the rat model of OAT after intragastric administration of adenine was selected as the research object, and randomly divided into a blank group (C), a model group (M), and a electroacupuncture therapy with distant-approximal acupoints group (D). After electroacupuncture intervention, the epididymal sperm quality and serum sex hormone levels of rats was detected and three tissue samples of HPT axis were taken, and differentially expressed genes (DEGs) were screened by transcriptome sequencing technology. GO functional annotation and KEGG pathway enrichment analysis were performed on the DEGs. The oxidative stress related indicators in serum and HPT axis were also detected to verify the transcriptomic analysis results. Compared with group C, group M rats showed a decrease in sperm count (p < 0.001), sperm survival rate (p < 0.001), and sperm motility rate (p < 0.001); the serum levels of GnRH in the group M rats decreased (p < 0.001), FSH increased (p < 0.001), LH increased (p < 0.001), and T decreased (p < 0.001). Compared with group M, group D rats showed an increase in sperm count (p < 0.01), sperm survival rate (p < 0.001), sperm motility rate (p < 0.001), an increase in GnRH levels (p < 0.001), a decrease in FSH levels (p < 0.01), a decrease in LH levels (p < 0.001), and an increase in T levels (p < 0.001). In bioinformatics analysis, compared with group M, we identified 1656, 518, 530 DEGs in the hypothalamus, pituitary, and testis in group D, respectively. Combining the go and KEGG analysis results, three oxidative stress signaling pathways that may be related to electroacupuncture intervention in OAT rats were screened. It mainly involves the glutamatergic synaptic pathway, the MAPK signaling pathway and the glutathione metabolism pathway. Six key genes (Gng12、Grin1、Gng7、Jun、Nf1 and Gstp1) were identified as key candidate genes regulating epididymal sperm quality on the HPT axis, which may affect the reproductive function of rats by affecting the process of oxidative stress in vivo. No matter in serum or in three tissues of HPT axis, GPX4 level in group M was decreased compared with Group K (p < 0.0001), while GPX4 level in group D was increased compared with group M (p < 0.0001). This study found that the effect of electroacupuncture therapy with distant-approximal acupoints based on the HPT axis in rats with OAT is related to the process of oxidative stress. And the main genes involved in the oxidative stress pathway were identified, which provided directions and ideas for subsequent research. But these results are only the preliminary results of transcriptomics, and relevant experiments need to be designed to further verify the mechanism of electroacupuncture therapy in rats with OAT.
Collapse
Affiliation(s)
- Li Hongyu
- Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
- General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
- Ningxia Regional Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Regional High Incidence Disease, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Yang Nan
- Ningxia Health Vocational and Technical College, Shizuishan, 753000, Ningxia Hui Autonomous Region, China
| | - Li Kaiying
- Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
- Ningxia Regional Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Regional High Incidence Disease, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Zhao Zhenning
- Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
- Ningxia Regional Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Regional High Incidence Disease, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Zhao Lili
- Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
- Ningxia Regional Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Regional High Incidence Disease, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Mu Jing
- Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China.
| | - Ma Huisheng
- Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China.
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, 750004, Ningxia Hui Autonomous Region, China.
- Ningxia Regional Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Regional High Incidence Disease, Yinchuan, 750004, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
6
|
Jung ES, Choi H, Mook-Jung I. Decoding microglial immunometabolism: a new frontier in Alzheimer's disease research. Mol Neurodegener 2025; 20:37. [PMID: 40149001 PMCID: PMC11948825 DOI: 10.1186/s13024-025-00825-0] [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: 09/01/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Alzheimer's disease (AD) involves a dynamic interaction between neuroinflammation and metabolic dysregulation, where microglia play a central role. These immune cells undergo metabolic reprogramming in response to AD-related pathology, with key genes such as TREM2, APOE, and HIF-1α orchestrating these processes. Microglial metabolism adapts to environmental stimuli, shifting between oxidative phosphorylation and glycolysis. Hexokinase-2 facilitates glycolytic flux, while AMPK acts as an energy sensor, coordinating lipid and glucose metabolism. TREM2 and APOE regulate microglial lipid homeostasis, influencing Aβ clearance and immune responses. LPL and ABCA7, both associated with AD risk, modulate lipid processing and cholesterol transport, linking lipid metabolism to neurodegeneration. PPARG further supports lipid metabolism by regulating microglial inflammatory responses. Amino acid metabolism also contributes to microglial function. Indoleamine 2,3-dioxygenase controls the kynurenine pathway, producing neurotoxic metabolites linked to AD pathology. Additionally, glucose-6-phosphate dehydrogenase regulates the pentose phosphate pathway, maintaining redox balance and immune activation. Dysregulated glucose and lipid metabolism, influenced by genetic variants such as APOE4, impair microglial responses and exacerbate AD progression. Recent findings highlight the interplay between metabolic regulators like REV-ERBα, which modulates lipid metabolism and inflammation, and Syk, which influences immune responses and Aβ clearance. These insights offer promising therapeutic targets, including strategies aimed at HIF-1α modulation, which could restore microglial function depending on disease stage. By integrating metabolic, immune, and genetic factors, this review underscores the importance of microglial immunometabolism in AD. Targeting key metabolic pathways could provide novel therapeutic strategies for mitigating neuroinflammation and restoring microglial function, ultimately paving the way for innovative treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- Eun Sun Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Hayoung Choi
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Inhee Mook-Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.
- Korea Dementia Research Center, Seoul, South Korea.
| |
Collapse
|
7
|
Fry LG, Washam CL, Roys H, Bowlin AK, Venugopal G, Bird JT, Byrum SD, Weinkopff T. HIF-α signaling regulates the macrophage inflammatory response during Leishmania major infection. Front Immunol 2025; 16:1487311. [PMID: 40191198 PMCID: PMC11969800 DOI: 10.3389/fimmu.2025.1487311] [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: 08/27/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Cutaneous leishmaniasis (CL) contributes significantly to the global burden of neglected tropical diseases, with 12 million people currently infected with Leishmania parasites. CL encompasses a range of disease manifestations, from self-healing skin lesions to permanent disfigurations. Currently there is no vaccine available, and many patients are refractory to treatment, emphasizing the need for new therapeutic targets. Previous work demonstrated macrophage HIF-α-mediated lymphangiogenesis is necessary to achieve efficient wound resolution during murine L. major infection. Here, we investigate the role of macrophage HIF-α signaling independent of lymphangiogenesis. We sought to determine the relative contributions of the parasite and the host-mediated inflammation in the lesional microenvironment to myeloid HIF-α signaling. Because HIF-α activation can be detected in infected and bystander macrophages in leishmanial lesions, we hypothesize it is the host's inflammatory response and microenvironment, rather than the parasite, that triggers HIF-α activation. To address this, macrophages from mice with intact HIF-α signaling (LysMCreARNTf/+) or mice with deleted HIF-α signaling (LysMCreARNTf/f) were subjected to RNASequencing after L. major infection and under pro-inflammatory stimulus. We report that L. major infection alone is enough to induce some minor HIF-α-dependent transcriptomic changes, while infection with L. major in combination with pro-inflammatory stimuli induces numerous transcriptomic changes that are both dependent and independent of HIF-α signaling. Additionally, by coupling transcriptomic analysis with several pathway analyses, we found HIF-α suppresses pathways involved in protein translation during L. major infection in a pro-inflammatory environment. Together these findings show L. major induces a HIF-α-dependent transcriptomic program, but HIF-α only suppresses protein translation in a pro-inflammatory environment. Thus, this work indicates the host inflammatory response, rather than the parasite, largely contributes to myeloid HIF-α signaling during Leishmania infection.
Collapse
Affiliation(s)
- Lucy G. Fry
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Charity L. Washam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - Hayden Roys
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Anne K. Bowlin
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Gopinath Venugopal
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jordan T. Bird
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
8
|
Heidari F, Kazemi-Sefat NA, Feizollahi P, Gerdabi S, Pourfathollah AA, Ebtekar M. Effect of FLT3 ligand on the gene expression of TIM-3, HIF1-α, and TNF-α in an acute myeloid leukemia cell line. Mol Biol Rep 2025; 52:313. [PMID: 40085277 DOI: 10.1007/s11033-025-10396-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Acute Myeloid Leukemia (AML) pathogenesis is driven by the dysregulation of various cell signaling pathways, including the FMS-Like Tyrosine Kinase 3 (FLT3) pathway and its ligand (FLT3L). These pathways play a critical role in promoting cell survival, proliferation, and resistance to apoptosis, contributing to leukemogenesis. In this study, we investigated the effects of FLT3L on the expression of key genes associated with immune regulation, hypoxia, and inflammation-TIM-3, HIF-1α, and TNF-α-in the THP-1 cell line, a well-established model for AML research. METHODS THP-1 cells were cultured under standard conditions and treated with varying concentrations of FLT3L, alongside PMA as a positive control. Quantitative RT-PCR was employed to measure the expression levels of TIM-3, HIF-1α, and TNF-α genes after 48 h of treatment. RESULTS Our findings demonstrated that specific concentrations of FLT3L significantly upregulated the expression of TIM-3, HIF-1α, and TNF-α in THP-1 cells. This suggests that FLT3L not only influences cell proliferation and survival but also modulates pathways related to immune evasion, hypoxia adaptation, and inflammatory responses, which are hallmarks of leukemia progression. CONCLUSION These results highlight the pivotal role of FLT3L in regulating the expression of genes associated with AML pathogenesis, particularly those involved in hypoxia (HIF-1α), immune checkpoint regulation (TIM-3), and inflammation (TNF-α). The findings underscore the potential of targeting the FLT3 pathway as a therapeutic strategy in AML. Further studies are warranted to elucidate the underlying molecular mechanisms and explore their clinical implications for improving patient outcomes.
Collapse
MESH Headings
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
- Hepatitis A Virus Cellular Receptor 2/genetics
- Hepatitis A Virus Cellular Receptor 2/metabolism
- THP-1 Cells
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Cell Line, Tumor
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- Signal Transduction/genetics
- Signal Transduction/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Cell Survival/drug effects
- Cell Survival/genetics
Collapse
Affiliation(s)
- Fatemeh Heidari
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Nazanin Atieh Kazemi-Sefat
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Parisa Feizollahi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Sajjad Gerdabi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Ali Akbar Pourfathollah
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Masoumeh Ebtekar
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran.
| |
Collapse
|
9
|
Cabezón-Gutiérrez L, Palka-Kotlowska M, Custodio-Cabello S, Chacón-Ovejero B, Pacheco-Barcia V. Metabolic mechanisms of immunotherapy resistance. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002297. [PMID: 40092297 PMCID: PMC11907103 DOI: 10.37349/etat.2025.1002297] [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: 12/08/2024] [Accepted: 02/22/2025] [Indexed: 03/19/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment, yet its efficacy is frequently compromised by metabolic mechanisms that drive resistance. Understanding how tumor metabolism shapes the immune microenvironment is essential for developing effective therapeutic strategies. This review examines key metabolic pathways influencing immunotherapy resistance, including glucose, lipid, and amino acid metabolism. We discuss their impact on immune cell function and tumor progression, highlighting emerging therapeutic strategies to counteract these effects. Tumor cells undergo metabolic reprogramming to sustain proliferation, altering the availability of essential nutrients and generating toxic byproducts that impair cytotoxic T lymphocytes (CTLs) and natural killer (NK) cell activity. The accumulation of lactate, deregulated lipid metabolism, and amino acid depletion contribute to an immunosuppressive tumor microenvironment (TME). Targeting metabolic pathways, such as inhibiting glycolysis, modulating lipid metabolism, and restoring amino acid balance, has shown promise in enhancing immunotherapy response. Addressing metabolic barriers is crucial to overcoming immunotherapy resistance. Integrating metabolic-targeted therapies with immune checkpoint inhibitors may improve clinical outcomes. Future research should focus on personalized strategies to optimize metabolic interventions and enhance antitumor immunity.
Collapse
Affiliation(s)
- Luis Cabezón-Gutiérrez
- Medical Oncology, Hospital Universitario De Torrejón, 28850 Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Magda Palka-Kotlowska
- Medical Oncology, Hospital Universitario De Torrejón, 28850 Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Sara Custodio-Cabello
- Medical Oncology, Hospital Universitario De Torrejón, 28850 Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Beatriz Chacón-Ovejero
- Department of Pharmacy and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Vilma Pacheco-Barcia
- Medical Oncology, Hospital Universitario De Torrejón, 28850 Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| |
Collapse
|
10
|
Yang H, Chen Y, Dai C, Xing Y, Qiu Z, Zhao J, Ye J, Yu C, Lin P, Zhang W, Zhang L, Luan X. Huachansu suppresses colorectal cancer via inhibiting PI3K/AKT and glycolysis signaling pathways: Systems biology and network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119479. [PMID: 39938766 DOI: 10.1016/j.jep.2025.119479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/01/2025] [Accepted: 02/10/2025] [Indexed: 02/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huachansu (HCS), a traditional Chinese medicine (TCM), has been used as an adjuvant therapy for colorectal cancer (CRC). However, its underlying mechanisms for combating CRC require further investigation. AIM OF THIS STUDY To comprehensively evaluate the anti-CRC effects of HCS and elucidate its underlying mechanisms, with a focus on elucidating the key pathways and targets involved. MATERIALS AND METHODS A series of cell experiments and xenograft tumor models were used to evaluate the inhibitory effects of HCS. The key components and potential targets of HCS against CRC were identified through network pharmacology and molecular docking. To further investigate the mechanisms, transcriptomics and proteomics were integrated, and the findings were supported by systematic pharmacological validation. Finally, the efficacy of HCS was further confirmed in CRC Patients-derived organoid and orthotopic models. RESULTS HCS could inhibit proliferation, disrupt the cell cycle, induce apoptosis of CRC cells, and suppress the growth of CRC xenograft tumors. Then eight components and six proteins (PIK3CA, CTNNB1, TP53, AKT1, CCND1, and CDH1) were identified as critical for HCS's anti-CRC activity. Notably, HCS inhibited the PI3K/AKT signaling pathway and glycolysis in CRC cells, with these findings validated in both in vitro and in vivo models. Additionally, HCS reduced growth in CRC patient-derived organoids and orthotopic models. CONCLUSION This study elucidates the mechanisms of HCS to combat CRC, offering a valuable reference for future clinical applications. It also presents a distinctive strategy for exploring TCM formulations' active components and effective mechanisms.
Collapse
Affiliation(s)
- Hongxuan Yang
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yixu Chen
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chunlan Dai
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yizhuo Xing
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ziyang Qiu
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zhao
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ji Ye
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Chenhua Yu
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Pengfei Lin
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Shenzhen Traditional Chinese Medicine Manufacturing Innovation Center Co., Ltd., Shenzhen, 518110, China
| | - Weidong Zhang
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Naval Medical University, Shanghai, 200433, China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Lijun Zhang
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center of Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
11
|
Tuffs C, Dupovac M, Richter K, Holten S, Schaschinger T, Marg O, Poljo A, Tasdemir AN, Harnoss JM, Billeter A, Schneider M, Strowitzki MJ. Genetic Loss of HIF-Prolyl-Hydroxylase 1, but Not Pharmacological Inhibition, Mitigates Hepatic Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:480-493. [PMID: 39566823 DOI: 10.1016/j.ajpath.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024]
Abstract
Liver fibrosis is characterized by excessive deposition of extracellular matrix due to chronic inflammation of the liver. Hepatic stellate cells (HSCs) become activated and produce increased amounts of extracellular matrix. Loss of HIF-prolyl-hydroxylase 1 (PHD1) attenuates HSC activation and fibrotic tissue remodeling in a murine model of biliary liver fibrosis. Herein, the protective effect of PHD1 deficiency (PHD1-/-) in an additional (toxic) model of liver fibrosis was validated and the effect of dimethyloxalylglycine (DMOG), a pan-HIF-prolyl-hydroxylase inhibitor, on the development of liver fibrosis, was evaluated. Liver fibrosis was induced utilizing carbon tetrachloride in wild-type (WT) and PHD1-/- mice treated with either vehicle or DMOG. To assess fibrosis development, expression of profibrotic genes in the livers was analyzed by Sirius red staining. When compared with WT mice, PHD1-/- mice developed less-severe liver fibrosis. DMOG treatment did not prevent this liver fibrosis. PHD1-/- mice had fewer α-SMA+ cells and less macrophage infiltration compared with WT mice. Expression of profibrogenic and proinflammatory genes was reduced in livers from carbon tetrachloride-exposed PHD1-/- mice. In vitro analyses of PHD1-deficient human HSCs revealed attenuated mRNA levels of profibrotic genes, as well as impaired migration and invasion. Although PHD1 deficiency attenuated activation of HSCs, pharmacologic PHD inhibition did not ameliorate fibrosis development. These data indicate that selective PHD1 inhibitors could prove effective in preventing and treating liver fibrosis.
Collapse
Affiliation(s)
- Christopher Tuffs
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Mareen Dupovac
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Katrin Richter
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany; Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Sophia Holten
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Thomas Schaschinger
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Oliver Marg
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Adisa Poljo
- Clarunis University Digestive Healthcare Center Basel, Basel, Switzerland
| | - Ayse Nur Tasdemir
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Adrian Billeter
- Clarunis University Digestive Healthcare Center Basel, Basel, Switzerland
| | - Martin Schneider
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Moritz J Strowitzki
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany.
| |
Collapse
|
12
|
Suvac A, Ashton J, Bristow RG. Tumour hypoxia in driving genomic instability and tumour evolution. Nat Rev Cancer 2025; 25:167-188. [PMID: 39875616 DOI: 10.1038/s41568-024-00781-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/30/2025]
Abstract
Intratumour hypoxia is a feature of all heterogenous solid tumours. Increased levels or subregions of tumour hypoxia are associated with an adverse clinical prognosis, particularly when this co-occurs with genomic instability. Experimental evidence points to the acquisition of DNA and chromosomal alterations in proliferating hypoxic cells secondary to inhibition of DNA repair pathways such as homologous recombination, base excision repair and mismatch repair. Cell adaptation and selection in repair-deficient cells give rise to a model whereby novel single-nucleotide mutations, structural variants and copy number alterations coexist with altered mitotic control to drive chromosomal instability and aneuploidy. Whole-genome sequencing studies support the concept that hypoxia is a critical microenvironmental cofactor alongside the driver mutations in MYC, BCL2, TP53 and PTEN in determining clonal and subclonal evolution in multiple tumour types. We propose that the hypoxic tumour microenvironment selects for unstable tumour clones which survive, propagate and metastasize under reduced immune surveillance. These aggressive features of hypoxic tumour cells underpin resistance to local and systemic therapies and unfavourable outcomes for patients with cancer. Possible ways to counter the effects of hypoxia to block tumour evolution and improve treatment outcomes are described.
Collapse
Affiliation(s)
- Alexandru Suvac
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jack Ashton
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert G Bristow
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK.
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK.
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
13
|
Espín R, Medina-Jover F, Sigüenza-Andrade J, Farran-Matas S, Mateo F, Figueras A, Sanz R, Vicent G, Shabbir A, Ruiz-Auladell L, Racionero-Andrés E, García I, Baiges A, Franco-Luzón L, Martínez-Tebar A, Pardo-Cea M, Martínez-Iniesta M, Wang X, Cuyàs E, Menendez J, Lopez-Cerda M, Muñoz P, Richaud I, Raya A, Fabregat I, Villanueva A, Serrat X, Cerón J, Alemany M, Guix I, Herencia-Ropero A, Serra V, Krishnan R, Mekhail K, Hakem R, Bruna J, Barcellos-Hoff M, Viñals F, Aytes Á, Pujana M. Harnessing transcriptional regulation of alternative end-joining to predict cancer treatment. NAR Cancer 2025; 7:zcaf007. [PMID: 40061566 PMCID: PMC11886861 DOI: 10.1093/narcan/zcaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/13/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
Alternative end-joining (alt-EJ) is an error-prone DNA repair pathway that cancer cells deficient in homologous recombination rely on, making them vulnerable to synthetic lethality via inhibition of poly(ADP-ribose) polymerase (PARP). Targeting alt-EJ effector DNA polymerase theta (POLθ), which synergizes with PARP inhibitors and can overcome resistance, is of significant preclinical and clinical interest. However, the transcriptional regulation of alt-EJ and its interactions with processes driving cancer progression remain poorly understood. Here, we show that alt-EJ is suppressed by hypoxia while positively associated with MYC (myelocytomatosis oncogene) transcriptional activity. Hypoxia reduces PARP1 and POLQ expression, decreases MYC binding at their promoters, and lowers PARylation and alt-EJ-mediated DNA repair in cancer cells. Tumors with HIF1A mutations overexpress the alt-EJ gene signature. Inhibition of hypoxia-inducible factor 1α or HIF1A expression depletion, combined with PARP or POLθ inhibition, synergistically reduces the colony-forming capacity of cancer cells. Deep learning reveals the anticorrelation between alt-EJ and hypoxia across regions in tumor images, and the predictions for these and MYC activity achieve area under the curve values between 0.70 and 0.86. These findings further highlight the critical role of hypoxia in modulating DNA repair and present a strategy for predicting and improving outcomes centered on targeting alt-EJ.
Collapse
Affiliation(s)
- Roderic Espín
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Ferran Medina-Jover
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Department of Physiological Sciences, University of Barcelona, L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Javier Sigüenza-Andrade
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Sònia Farran-Matas
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Francesca Mateo
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Agnes Figueras
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Rosario T Sanz
- Molecular Biology Institute of Barcelona, Spanish National Research Council (IBMB-CSIC), Barcelona 08028, Spain
| | - Guillermo Pablo Vicent
- Molecular Biology Institute of Barcelona, Spanish National Research Council (IBMB-CSIC), Barcelona 08028, Spain
| | - Arzoo Shabbir
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Lara Ruiz-Auladell
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | | | - Irene García
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Alexandra Baiges
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Lídia Franco-Luzón
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Adrián Martínez-Tebar
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Miguel Angel Pardo-Cea
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - María Martínez-Iniesta
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Xieng Chen Wang
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Elisabet Cuyàs
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Girona Biomedical Research Institute (IDIBGI), Salt, Girona 17190, Spain
| | - Javier A Menendez
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Girona Biomedical Research Institute (IDIBGI), Salt, Girona 17190, Spain
| | - Marta Lopez-Cerda
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Purificacion Muñoz
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Ivonne Richaud
- Regenerative Medicine Program and Program for Clinical Translation of Regenerative Medicine in Catalonia—P-CMR[C], Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Biomedical Research Network Centre in Bioengineering, Nanomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Angel Raya
- Regenerative Medicine Program and Program for Clinical Translation of Regenerative Medicine in Catalonia—P-CMR[C], Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Biomedical Research Network Centre in Bioengineering, Nanomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08010, Spain
| | - Isabel Fabregat
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Biomedical Research Networking Centre in Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Alberto Villanueva
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Xènia Serrat
- Modeling Human Diseases in C. elegans Group, Genes, Diseases, and Therapies Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Julián Cerón
- Modeling Human Diseases in C. elegans Group, Genes, Diseases, and Therapies Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Montserrat Alemany
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Neuro-Oncology Unit, University Hospital of Bellvitge, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Inés Guix
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco, San Francisco, CA 94115, United States
| | - Andrea Herencia-Ropero
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona 08193, Spain
- Experimental Therapeutics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona 08035, Spain
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona 08035, Spain
| | - Rehna Krishnan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Razqallah Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jordi Bruna
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Neuro-Oncology Unit, University Hospital of Bellvitge, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco, San Francisco, CA 94115, United States
| | - Francesc Viñals
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Department of Physiological Sciences, University of Barcelona, L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Álvaro Aytes
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Miquel Angel Pujana
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Girona Biomedical Research Institute (IDIBGI), Salt, Girona 17190, Spain
| |
Collapse
|
14
|
Lee J, Roh JL. Ferroptosis: iron release mechanisms in the bioenergetic process. Cancer Metastasis Rev 2025; 44:36. [PMID: 40000477 DOI: 10.1007/s10555-025-10252-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
Ferroptosis, an iron-dependent form of cell death, has been the focus of extensive research over the past decade, leading to the elucidation of key molecules and mechanisms involved in this process. While several studies have highlighted iron sources for the Fenton reaction, the predominant mechanism for iron release in ferroptosis has been identified as ferritinophagy, which occurs in response to iron starvation. However, much of the existing literature has concentrated on lipid peroxidation rather than on the mechanisms of iron release. This review proposes three distinct mechanisms of iron mobilization: ferritinophagy, reductive pathways with selective gating of ferritin pores, and quinone-mediated iron mobilization. Notably, the latter two mechanisms operate independently of iron starvation and rely primarily on reductants such as NADH and O2•-. The inhibition of the respiratory chain, particularly under the activation of α-ketoglutarate dehydrogenase, leads to the accumulation of these reductants, which in turn promotes iron release from ferritin and indirectly inhibits AMP-activated protein kinase through excessive iron levels. In this work, we delineate the intricate relationship between iron mobilization and bioenergetic processes under conditions of oxidative stress. Furthermore, this review aims to enhance the understanding of the connections between ferroptosis and these mechanisms.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea
- Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea.
- Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea.
| |
Collapse
|
15
|
Daumova L, Manakov D, Petrak J, Sovilj D, Behounek M, Andera L, Vit O, Souckova O, Havranek O, Dolnikova A, Renesova N, Tuskova L, Winkowska L, Bettazova N, Kupcova K, Kalbacova MH, Sikorova M, Trneny M, Klener P. Long-term adaptation of lymphoma cell lines to hypoxia is mediated by diverse molecular mechanisms that are targetable with specific inhibitors. Cell Death Discov 2025; 11:65. [PMID: 39966387 PMCID: PMC11836139 DOI: 10.1038/s41420-025-02341-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
A large body of evidence suggests that hypoxia drives aggressive molecular features of malignant cells irrespective of cancer type. Non-Hodgkin lymphomas (NHL) are the most common hematologic malignancies characterized by frequent involvement of diverse hypoxic microenvironments. We studied the impact of long-term deep hypoxia (1% O2) on the biology of lymphoma cells. Only 2 out of 6 tested cell lines (Ramos, and HBL2) survived ≥ 4 weeks under hypoxia. The hypoxia-adapted (HA)b Ramos and HBL2 cells had a decreased proliferation rate accompanied by significant suppression of both oxidative phosphorylation and glycolytic pathways. Transcriptome and proteome analyses revealed marked downregulation of genes and proteins of the mitochondrial respiration complexes I and IV, and mitochondrial ribosomal proteins. Despite the observed suppression of glycolysis, the proteome analysis of both HA cell lines showed upregulation of several proteins involved in the regulation of glucose utilization including the active catalytic component of prolyl-4-hydroxylase P4HA1, an important druggable oncogene. HA cell lines demonstrated increased transcription of key regulators of auto-/mitophagy, e.g., neuritin, BCL2 interacting protein 3 (BNIP3), BNIP3-like protein, and BNIP3 pseudogene. Adaptation to hypoxia was further associated with deregulation of apoptosis, namely upregulation of BCL2L1/BCL-XL, overexpression of BCL2L11/BIM, increased binding of BIM to BCL-XL, and significantly increased sensitivity of both HA cell lines to A1155463, a BCL-XL inhibitor. Finally, in both HA cell lines AKT kinase was hyperphosphorylated and the cells showed increased sensitivity to copanlisib, a pan-PI3K inhibitor. In conclusion, our data report on several shared mechanisms of lymphoma cell adaptation to long-term hypoxia including: 1. Upregulation of proteins responsible for glucose utilization, 2. Degradation of mitochondrial proteins for potential mitochondrial recycling (by mitophagy), and 3. Increased dependence on BCL-XL and PI3K-AKT signaling for survival. In translation, inhibition of glycolysis, BCL-XL, or PI3K-AKT cascade may result in targeted elimination of HA lymphoma cells.
Collapse
Affiliation(s)
- Lenka Daumova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Dmitry Manakov
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiri Petrak
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Dana Sovilj
- Institute of Biotechnology, Czech Academy of Sciences / BIOCEV, Vestec, Czech Republic
| | - Matej Behounek
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Ladislav Andera
- Institute of Biotechnology, Czech Academy of Sciences / BIOCEV, Vestec, Czech Republic
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Ondrej Vit
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Olga Souckova
- OMICS Mass Spectrometry Core Facility, Biology Departments, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Ondrej Havranek
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alex Dolnikova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Nicol Renesova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Liliana Tuskova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lucie Winkowska
- CLIP, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Nardjas Bettazova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kristyna Kupcova
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marie Hubalek Kalbacova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Miriama Sikorova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marek Trneny
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
16
|
Ferdousmakan S, Mansourian D, Seyedi Asl FS, Fathi Z, Maleki-Sheikhabadi F, Afjadi MN, Zalpoor H. Autophagy induced by metabolic processes leads to solid tumor cell metastatic dormancy and recurrence. Med Oncol 2025; 42:62. [PMID: 39899220 DOI: 10.1007/s12032-025-02607-6] [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: 10/22/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
A crucial cellular mechanism that has a complex impact on the biology of cancer, particularly in solid tumors, is autophagy. This review explores how metabolic processes trigger autophagy, which helps metastatic tumor cells go dormant and recur. During metastasis, tumor cells frequently encounter severe stressors, such as low oxygen levels and nutritional deprivation, which causes them to activate autophagy as a survival tactic. This process allows cancer stem cells (CSCs) to withstand severe conditions while also preserving their features. After years of dormancy, dormant disseminated tumor cells (DTCs) may reappear as aggressive metastatic cancers. The capacity of autophagy to promote resistance to treatments and avoid immune detection is intimately related to this phenomenon. According to recent research, autophagy promotes processes, such as the epithelial-to-mesenchymal transition (EMT) and helps build a pre-metastatic niche, which makes treatment strategies more challenging. Autophagy may be a promising therapeutic target because of its dual function as a tumor suppressor in early-stage cancer and a survival promoter in advanced stages. To effectively treat metastatic diseases, it is crucial to comprehend how metabolic processes interact with autophagy and affect tumor behavior. In order to find novel therapeutic approaches that can interfere with these processes and improve patient outcomes, this study highlights the critical need for additional investigation into the mechanisms by which autophagy controls tumor dormancy and recurrence.
Collapse
Affiliation(s)
- Saeid Ferdousmakan
- Department of Pharmacy Practice, Nargund College of Pharmacy, Bangalore, 560085, India
| | - Dorrin Mansourian
- Faculty of Pharmacy, Eastern Mediterranean University, Gazimagusa TRNC via Mersin 10, Mersin, Turkey
| | | | - Zeinab Fathi
- Medical School, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
17
|
Mao C, Liu X, Guo SW. Reduced endometrial glycolysis concomitant with increased lesional fibrosis in patients with adenomyosis who complained of heavy menstrual bleeding. Reprod Biomed Online 2025; 50:104406. [PMID: 39523182 DOI: 10.1016/j.rbmo.2024.104406] [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: 04/27/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 11/16/2024]
Abstract
RESEARCH QUESTION What role, if any, does the extent of lesional fibrosis play in impaired glycolysis leading to adenomyosis-associated heavy menstrual bleeding (ADM-HMB)? DESIGN Forty-eight patients with ADM-HMB were recruited, among them 25 reported moderate to heavy bleeding (MHB), and the remaining 23, excessive bleeding (EXB). The full-thickness uterine tissue columns were processed for Masson trichrome staining and immunohistochemistry analyses. The expression levels of HIF-1α, GLUT1, HK2, PFKFB3 and PKM2 proteins that are critically involved in glycolysis in endometrial epithelial cells cultured on substrates of different stiffness, and the levels of glycolysis were quantitated. A mouse experiment with induced adenomyosis and simulated menstrual bleeding was conducted to assess the effect of adenomyosis on immunoexpression of proteins involved in glycolysis and inflammation as well as on endometrial repair and bleeding. RESULTS The endometrial staining of HIF-1α, GLUT1, HK2, PFKFB3 and PKM2 was significantly lower in the EXB group as compared with MHB patients, concomitant with higher extent of fibrosis. The expression of HIF-1α, GLUT1, HK2, PFKFB3 and PKM2 was significantly reduced when endometrial epithelial cells were cultured in stiff substrate, concomitant with reduced glycolysis. Mice with induced adenomyosis had reduced immunoexpression of Hif-1α, as well as those proteins each of which plays a vital, rate-limiting role in different steps of the glycolysis pathway, such as Glut1, Hk2, Pfkfb3 and Pkm2, and elevated fibrosis in endometrium, concomitant with disrupted endometrial repair and more bleeding. CONCLUSIONS Lesional fibrosis results in reduced endometrial glycolysis in eutopic endometrium and subsequent imbalance in pro-inflammatory and anti-inflammatory response, leading to ADM-HMB.
Collapse
Affiliation(s)
- Chenyu Mao
- Department of Gynecology, Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Xishi Liu
- Department of Gynecology, Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China.; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| | - Sun-Wei Guo
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China.; Research Institute, Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China.
| |
Collapse
|
18
|
Zhao T, Cai X, Chen H, Wang Z, Bu H, Lin S. Rg3 inhibits hypoxia-induced tumor exosomes from boosting pancreatic cancer vasculogenic mimicry through the HIF-1α/LARS1/mTOR axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156437. [PMID: 39955826 DOI: 10.1016/j.phymed.2025.156437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 01/15/2025] [Accepted: 01/27/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Pancreatic cancer (PC), characterized by a poor prognosis, can utilize hypoxia to activate vasculogenic mimicry (VM) and facilitate metastasis. Exosomes serve as crucial mediators in this hypoxic environment. Our previous studies demonstrated that Rg3 could counteract VM by modulating the impact of PC-derived exosomes. AIM This study focuses on the carcinogenic mechanism of PC in a hypoxic environment. METHODS Exosomes from PANC-1 and BxPC-3 cells under normoxic or hypoxic conditions were isolated and characterized by western blot (WB) and nanoparticle trafficking analysis. These PC cells' VM potential was assessed through tube formation and WB. Molecular mechanisms were explored using proteomics analysis, bioinformatics analysis, and the gain- and loss-of-function studies, and the efficacy of Rg3 targeting these exosomes was examined in vitro and in vivo. RESULTS Exosomes from PANC-1 and BxPC-3 cells enhanced VM formation in PC cells under hypoxic conditions. Proteomics analysis revealed that these exosomes involved the HIF-1α/LARS1/mTOR axis. Hypoxia-activated HIF-1α led to high expression of LARS1 in PC cell exosomes, which were uptaken by recipient PC cells activating the mTOR signaling and promoting VM formation. Interaction between HIF-1α and LARS1 was further confirmed. In vitro and in vivo experiments demonstrated that Rg3 can diminish VM formation of PC cells triggered by the LARS1/mTOR axis in PC-derived exosomes under hypoxic conditions, improving the therapeutic effect of Rg3. CONCLUSIONS Our findings revealed a novel mechanism through which Rg3 inhibits VM in PC by modulating hypoxia-induced tumor exosomes, offering novel experimental insights for PC treatment.
Collapse
Affiliation(s)
- Tingting Zhao
- School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, PR China
| | - Xufan Cai
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, Zhejiang, PR China; Cancer Center, Department of Thoracic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, Zhejiang, PR China
| | - Hui Chen
- Department of hepatobiliary and pancreatic surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Zhaohong Wang
- Department of hepatobiliary and pancreatic surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Heqi Bu
- Department of Colorectal Surgery, Beilun District People's Hospital, Ningbo 315800, Zhejiang, PR China
| | - Shengzhang Lin
- School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, PR China.
| |
Collapse
|
19
|
Gong L, Zou C, Zhang H, Yang F, Qi G, Ma Z. Landscape of Noncoding RNA in the Hypoxic Tumor Microenvironment. Genes (Basel) 2025; 16:140. [PMID: 40004471 PMCID: PMC11855738 DOI: 10.3390/genes16020140] [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: 01/06/2025] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Amidst the prevalent and notable characteristic of a hypoxic microenvironment present in the majority of solid tumors, a burgeoning number of studies have revealed the significance of noncoding RNAs (ncRNAs) in hypoxic tumor regions. The transcriptome of cancers is highly heterogeneous, with noncoding transcripts playing crucial roles. Long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) are two distinctive classes of ncRNA that are garnering increasing attention. Biologically, they possess intriguing properties and possess significant regulatory functions. Clinically, they present as promising biomarkers and therapeutic targets. Additionally, recent research has evaluated the clinical applications of these ncRNAs in RNA-based treatments and noninvasive liquid biopsies. This review provides a comprehensive summary of recent studies on lncRNAs and circRNAs within the hypoxic tumor microenvironment. Furthermore, the clinical significance of lncRNAs and circRNAs in cancer diagnosis and treatment is emphasized, which could pave the way for the development of effective targeted therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (L.G.); (C.Z.); (H.Z.); (F.Y.); (G.Q.)
| |
Collapse
|
20
|
Agarwal AP, Kumar MS. Effect of epigenetic changes in hypoxia induced factor (HIF) gene across cancer types. Gene 2025; 934:149047. [PMID: 39490706 DOI: 10.1016/j.gene.2024.149047] [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: 05/17/2024] [Revised: 09/23/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Cancer hypoxia, a crucial characteristic of malignancy, ranging from practically non-hypoxic to severe, impacts gene expression, metabolism and mechanisms associated with tumor formation serves as a key obstacle in cancer therapy. It triggers a complex network of cell signaling pathways, such as the NF-κB, PI3K, mTOR/AKT,MAPK, HIF and their associated genes regulating the effects of the same. The onset and advancement of cancer are attributed to genetic and epigenetic modifications which are intrinsically related. Off late, it has been observed that in disease progression, the epigenetic modifications lead to gene mutations that in turn alter the epigenome, presenting a major hurdle in fabricating treatment strategies. However, theprogress in science and technology has led to the emergence of various surfacing omics and multi-view clustering algorithms, which offer unparalleled prospects for further subtyping cancers, enhancing the prognosis and treatment results of these subtypes, and comprehending crucial pathophysiological mechanisms across diverse molecular strata. Multi-omics has allowed scientists to gain a more comprehensive understanding of the various ways that cellular malfunction can lead to cancer. So, it becomes of utmost importance to firstly understand the epigenetic changes taking place in tumor hypoxia at gene level. This review sheds light on the role of HIF gene in hypoxic milieu and its relationship with mechanisms of cancer epigenetics. It further glances as to how omics approach can be used to study the oncogenic cellular changes and how bioinformatic tools aid in identification of complex gene networks involved in disease progression. Lastly, it glimpses through the benefits and shortcomings of the existing epi drug therapy and how it can be used in developing novel treatment options.
Collapse
Affiliation(s)
- Aditi P Agarwal
- Somaiya Institute for Research and Consultancy, Somaiya Vidyavihar University, Vidyavihar (East), Mumbai 400077, India
| | - Maushmi S Kumar
- Somaiya Institute for Research and Consultancy, Somaiya Vidyavihar University, Vidyavihar (East), Mumbai 400077, India..
| |
Collapse
|
21
|
Shi Y, Gilkes DM. HIF-1 and HIF-2 in cancer: structure, regulation, and therapeutic prospects. Cell Mol Life Sci 2025; 82:44. [PMID: 39825916 PMCID: PMC11741981 DOI: 10.1007/s00018-024-05537-0] [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: 08/05/2024] [Revised: 10/27/2024] [Accepted: 12/01/2024] [Indexed: 01/20/2025]
Abstract
Hypoxia, or a state of low tissue oxygenation, has been characterized as an important feature of solid tumors that is related to aggressive phenotypes. The cellular response to hypoxia is controlled by Hypoxia-inducible factors (HIFs), a family of transcription factors. HIFs promote the transcription of gene products that play a role in tumor progression including proliferation, angiogenesis, metastasis, and drug resistance. HIF-1 and HIF-2 are well known and widely described. Although these proteins share a high degree of homology, HIF-1 and HIF-2 have non-redundant roles in cancer. In this review, we summarize the similarities and differences between HIF-1α and HIF-2α in their structure, expression, and DNA binding. We also discuss the canonical and non-canonical regulation of HIF-1α and HIF-2α under hypoxic and normal conditions. Finally, we outline recent strategies aimed at targeting HIF-1α and/or HIF-2α.
Collapse
Affiliation(s)
- Yi Shi
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniele M Gilkes
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Ewida H, Benson H, Tareq S, Ahmed MS. Molecular Targets and Small Molecules Modulating Acetyl Coenzyme A in Physiology and Diseases. ACS Pharmacol Transl Sci 2025; 8:36-46. [PMID: 39816789 PMCID: PMC11729435 DOI: 10.1021/acsptsci.4c00476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
Acetyl coenzyme A (acetyl-CoA), a pivotal regulatory metabolite, is a product of numerous catabolic reactions and a substrate for various anabolic responses. Its role extends to crucial physiological processes, such as glucose homeostasis and free fatty acid utilization. Moreover, acetyl-CoA plays a significant part in reshaping the metabolic microenvironment and influencing the progression of several diseases and conditions, including cancer, insulin resistance, diabetes, heart failure, fear, and neuropathic pain. This Review delves into the role of acetyl-CoA in both physiological and pathological conditions, shedding light on the key players in its formation within the cytosol. We specifically focus on the physiological impact of malonyl-CoA decarboxylase (MCD), acetyl-CoA synthetase2 (ACSS2), and ATP-citrate lyase (ACLY) on metabolism, glucose homeostasis, free fatty acid utilization, and post-translational modification cellular processes. Additionally, we present the pathological implications of MCD, ACSS2, and ACLY in various clinical manifestations. This Review also explores the potential and limitations of targeting MCD, ACSS2, and ACLY using small molecules in different clinical settings.
Collapse
Affiliation(s)
- Heba Ewida
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, Texas 79106, United States
- Department
of Biochemistry, Faculty of Pharmacy, Future
University in Egypt, Cairo 11835, Egypt
| | - Harrison Benson
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, Texas 79106, United States
| | - Syed Tareq
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, Texas 79106, United States
| | - Mahmoud Salama Ahmed
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, Texas 79106, United States
| |
Collapse
|
23
|
Xu W, Huang X, Wu X. The effect of acute hypoxic exercise on the protein kinase A/ arachidonic acid/ transient receptor potential vanilloid 4 pathway in the prefrontal cortex of rats. Arch Biochem Biophys 2025; 763:110214. [PMID: 39566673 DOI: 10.1016/j.abb.2024.110214] [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: 07/27/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Acute hypoxic exercise will cause insufficient oxygen supply in brain tissue, and a succession of variations such as central dysfunction will occur. For example, the muscles don't have an adequate supply of oxygen, which leads to decrease in exercise capacity (Imray et al., 2005) [1]. The prefrontal cortex in the brain is primarily responsible for regulating the executive functions of the brain. TRPV4 channel is a cation channel with permeability to Ca2+. Signals such as hypotonic solution stimulation, cell swelling, temperature stimulation, mechanical stimulation, arachidonic acid and its metabolites can activate TRPV4 channel. PURPOSE In conditions of ischemia and hypoxia, the central nervous system of the brain is damaged. Therefore, studying the biological mechanism of TRPV4 pathway can help prevent the damage caused by cerebral ischemia and hypoxia to the human. RESULTS Studies have found that PKA-mediated phosphorylation at Ser-824 affects AA. This is an important signaling pathway for coronary dilatation. This signaling pathway can activate TRPV4 channels. Therefore, we studied the effect of acute hypoxic exercise on the PKA/AA/TRPV4 pathway in the prefrontal cortex of rats. Furthermore, we concluded that the hypoxic environment can shorten the time of increasing load exercise in rats. In this way, the rat entered a state of exhaustion in advance, and the exercise ability was significantly reduced. After further study, blocking TRPV4 channel can prolong the time of incremental load exercise in hypoxic environment, and the exercise ability is improved. Acute hypoxic exercise led to an increase in the concentration of 14,15-EET, which was speculated to be one of the reasons for the increased expression of TRPV4 channels. Acute hypoxic exercise can activate the PKA/AA/TRPV4 signaling pathway in the prefrontal cortex of rats. Further research on blocking the TRPV4 channel can alleviate the activation of the PKA/AA/TRPV4 signaling pathway in the prefrontal cortex of rats by acute hypoxic exercise. CONCLUSION These results suggest that blocking the TRPV4 channel may be one of the ways to reduce the damage and apoptosis of prefrontal cortex cells in rats due to acute hypoxic exercise. In future studies, multiple time points will be selected for collection. Alternatively, TRPV4 agonists, PKA agonists or blockers and 14,15-EET agonists or blockers can be added for further pathway validation. To provide a biological mechanism for the study of nutrient targets on the problem of reduced exercise capacity when military personnel and travel enthusiasts first went to the plateau. Better medical reference for athletes training at high altitudes or patients with respiratory issues.
Collapse
Affiliation(s)
- Wenlei Xu
- Capital University of Physical Education and Sports, Beijing, 100191, China
| | - Xing Huang
- Capital University of Physical Education and Sports, Beijing, 100191, China.
| | - Xiaolong Wu
- Capital University of Physical Education and Sports, Beijing, 100191, China
| |
Collapse
|
24
|
Song CW, Kim H, Kim MS, Park HJ, Paek SH, Terezakis S, Cho LC. Role of HIF-1α in the Responses of Tumors to Radiotherapy and Chemotherapy. Cancer Res Treat 2025; 57:1-10. [PMID: 38853541 PMCID: PMC11729307 DOI: 10.4143/crt.2024.255] [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: 03/12/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024] Open
Abstract
Tumor microenvironment is intrinsically hypoxic with abundant hypoxia-inducible factors-1α (HIF-1α), a primary regulator of the cellular response to hypoxia and various stresses imposed on the tumor cells. HIF-1α increases radioresistance and chemoresistance by reducing DNA damage, increasing repair of DNA damage, enhancing glycolysis that increases antioxidant capacity of tumors cells, and promoting angiogenesis. In addition, HIF-1α markedly enhances drug efflux, leading to multidrug resistance. Radiotherapy and certain chemotherapy drugs evoke profound anti-tumor immunity by inducing immunologic cell death that release tumor-associated antigens together with numerous pro-immunological factors, leading to priming of cytotoxic CD8+ T cells and enhancing the cytotoxicity of macrophages and natural killer cells. Radiotherapy and chemotherapy of tumors significantly increase HIF-1α activity in tumor cells. Unfortunately, HIF-1α effectively promotes various immune suppressive pathways including secretion of immune suppressive cytokines, activation of myeloid-derived suppressor cells, activation of regulatory T cells, inhibition of T cells priming and activity, and upregulation of immune checkpoints. Consequently, the anti-tumor immunity elevated by radiotherapy and chemotherapy is counterbalanced or masked by the potent immune suppression promoted by HIF-1α. Effective inhibition of HIF-1α may significantly increase the efficacy of radiotherapy and chemotherapy by increasing radiosensitivity and chemosensitivity of tumor cells and also by upregulating anti-tumor immunity.
Collapse
Affiliation(s)
- Chang W Song
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hyunkyung Kim
- Department of Radiation Oncology, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Mi-Sook Kim
- Department of Radiation Oncology, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Heon J Park
- Department of Microbiology, College of Medicine, Inha University, Incheon, Korea
| | - Sun-Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Korea
| | - Stephanie Terezakis
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - L Chinsoo Cho
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
25
|
Varshini MS, Krishnamurthy PT, Reddy RA, Wadhwani A, Chandrashekar VM. Insights into the Emerging Therapeutic Targets of Triple-negative Breast Cancer. Curr Cancer Drug Targets 2025; 25:3-25. [PMID: 38385495 DOI: 10.2174/0115680096280750240123054936] [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: 09/23/2023] [Revised: 12/24/2023] [Accepted: 01/09/2024] [Indexed: 02/23/2024]
Abstract
Triple-negative Breast Cancer (TNBC), the most aggressive breast cancer subtype, is characterized by the non-appearance of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Clinically, TNBC is marked by its low survival rate, poor therapeutic outcomes, high aggressiveness, and lack of targeted therapies. Over the past few decades, many clinical trials have been ongoing for targeted therapies in TNBC. Although some classes, such as Poly (ADP Ribose) Polymerase (PARP) inhibitors and immunotherapies, have shown positive therapeutic outcomes, however, clinical effects are not much satisfiable. Moreover, the development of drug resistance is the major pattern observed in many targeted monotherapies. The heterogeneity of TNBC might be the cause for limited clinical benefits. Hence,, there is a need for the potential identification of new therapeutic targets to address the above limitations. In this context, some novel targets that can address the above-mentioned concerns are emerging in the era of TNBC therapy, which include Hypoxia Inducible Factor (HIF-1α), Matrix Metalloproteinase 9 (MMP-9), Tumour Necrosis Factor-α (TNF-α), β-Adrenergic Receptor (β-AR), Voltage Gated Sodium Channels (VGSCs), and Cell Cycle Regulators. Currently, we summarize the ongoing clinical trials and discuss the novel therapeutic targets in the management of TNBC.
Collapse
Affiliation(s)
- Magham Sai Varshini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, 643001, TN, India
| | | | - Ramakamma Aishwarya Reddy
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, 643001, TN, India
| | - Ashish Wadhwani
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, 643001, TN, India
- Faculty of Health Sciences, School of Pharmacy, JSS Academy of Higher Education and Research, Mauritius, Vacoas, 73304, Mauritius
| | - V M Chandrashekar
- Department of Pharmacology, HSK College of Pharmacy, Bagalkot, 587101, Karnataka, India
| |
Collapse
|
26
|
Ni F, Wang F, Sun J, Tu M, Chen J, Shen X, Ye X, Chen R, Liu Y, Sun X, Chen J, Li X, Zhang D. Proteome-wide Mendelian randomization and functional studies uncover therapeutic targets for polycystic ovarian syndrome. Am J Hum Genet 2024; 111:2799-2813. [PMID: 39541979 PMCID: PMC11639085 DOI: 10.1016/j.ajhg.2024.10.008] [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: 08/01/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
Polycystic ovarian syndrome (PCOS) is an endocrine syndrome that affects a large portion of women worldwide. This proteogenomic and functional study aimed to uncover candidate therapeutic targets for PCOS. We comprehensively investigated the causal association between circulating proteins and PCOS using two-sample Mendelian randomization analysis. Cis-protein quantitative trait loci were derived from six genome-wide association studies (GWASs) on plasma proteome. Genetic associations with PCOS were obtained from a large-scale GWAS meta-analysis, FinnGen cohort, and UK Biobank. Colocalization analyses were performed to prioritize the causal role of candidate proteins. Protein-protein interaction (PPI) and druggability evaluation assessed the druggability of candidate proteins. We evaluated the enrichment of tier 1 and 2 candidate proteins in individuals with PCOS and a mouse model and explored the potential application of the identified drug target. Genetically predicted levels of 65 proteins exhibited associations with PCOS risk, with 30 proteins showing elevated levels and 35 proteins showing decreased levels linked to higher susceptibility. PPI analyses revealed that FSHB, POSTN, CCN2, and CXCL11 interacted with targets of current PCOS medications. Eighty medications targeting 20 proteins showed their potential for repurposing as therapeutic targets for PCOS. EGLN1 levels were elevated in granulosa cells and the plasma of individuals with PCOS and in the plasma and ovaries of dehydroepiandrosterone (DHEA)-induced PCOS mouse model. As an EGLN1 inhibitor, administration of roxadustat in the PCOS mouse model elucidated the EGLN1-HIF1α-ferroptosis axis in inducing PCOS and validated its therapeutic effect in PCOS. Our study identifies candidate proteins causally associated with PCOS risk and suggests that targeting EGLN1 provides a promising treatment strategy.
Collapse
Affiliation(s)
- Feida Ni
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China; First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Feixia Wang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Jing Sun
- Department of Big Data in Health Science, School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Mixue Tu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Jianpeng Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Xiling Shen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Xiaohang Ye
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Ruixue Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Yifeng Liu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Xiao Sun
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China
| | - Jianhua Chen
- Department of Pathology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Xue Li
- Department of Big Data in Health Science, School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Women's Reproductive Health Laboratory of Zhejiang Province, Hangzhou, Zhejiang 310006, China; Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
27
|
Cordani M, Michetti F, Zarrabi A, Zarepour A, Rumio C, Strippoli R, Marcucci F. The role of glycolysis in tumorigenesis: From biological aspects to therapeutic opportunities. Neoplasia 2024; 58:101076. [PMID: 39476482 PMCID: PMC11555605 DOI: 10.1016/j.neo.2024.101076] [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: 08/02/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 11/11/2024]
Abstract
Glycolytic metabolism generates energy and intermediates for biomass production. Tumor-associated glycolysis is upregulated compared to normal tissues in response to tumor cell-autonomous or non-autonomous stimuli. The consequences of this upregulation are twofold. First, the metabolic effects of glycolysis become predominant over those mediated by oxidative metabolism. Second, overexpressed components of the glycolytic pathway (i.e. enzymes or metabolites) acquire new functions unrelated to their metabolic effects and which are referred to as "moonlighting" functions. These functions include induction of mutations and other tumor-initiating events, effects on cancer stem cells, induction of increased expression and/or activity of oncoproteins, epigenetic and transcriptional modifications, bypassing of senescence and induction of proliferation, promotion of DNA damage repair and prevention of DNA damage, antiapoptotic effects, inhibition of drug influx or increase of drug efflux. Upregulated metabolic functions and acquisition of new, non-metabolic functions lead to biological effects that support tumorigenesis: promotion of tumor initiation, stimulation of tumor cell proliferation and primary tumor growth, induction of epithelial-mesenchymal transition, autophagy and metastasis, immunosuppressive effects, induction of drug resistance and effects on tumor accessory cells. These effects have negative consequences on the prognosis of tumor patients. On these grounds, it does not come to surprise that tumor-associated glycolysis has become a target of interest in antitumor drug discovery. So far, however, clinical results with glycolysis inhibitors have fallen short of expectations. In this review we propose approaches that may allow to bypass some of the difficulties that have been encountered so far with the therapeutic use of glycolysis inhibitors.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Federica Michetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, Milan 20134, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy.
| | - Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, Milan 20134, Italy.
| |
Collapse
|
28
|
Procházková K, Uhlík J. Influence of Hypoxia on the Airway Epithelium. Physiol Res 2024; 73:S557. [PMID: 39589303 PMCID: PMC11627265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 06/26/2024] [Indexed: 11/27/2024] Open
Abstract
The necessity of oxygen for metabolic processes means that hypoxia can lead to serious cell and tissue damage. On the other hand, in some situations, hypoxia occurs under physiological conditions and serves as an important regulation factor. The airway epithelium is specific in that it gains oxygen not only from the blood supply but also directly from the luminal air. Many respiratory diseases are associated with airway obstruction or excessive mucus production thus leading to luminal hypoxia. The main goal of this review is to point out how the airway epithelium reacts to hypoxic conditions. Cells detect low oxygen levels using molecular mechanisms involving hypoxia-inducible factors (HIFs). In addition, the cells of the airway epithelium appear to overexpress HIFs in hypoxic conditions. HIFs then regulate many aspects of epithelial cell functions. The effects of hypoxia include secretory cell stimulation and hyperplasia, epithelial barrier changes, and ciliogenesis impairment. All the changes can impair mucociliary clearance, exacerbate infection, and promote inflammation leading to damage of airway epithelium and subsequent airway wall remodeling. The modulation of hypoxia regulatory mechanisms may be one of the strategies for the treatment of obstructive respiratory diseases or diseases with mucus hyperproduction. Keywords: Secretory cells, Motile cilia, Epithelial barrier, Oxygenation, Obstructive respiratory diseases.
Collapse
Affiliation(s)
- K Procházková
- Department of Histology and Embryology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | |
Collapse
|
29
|
Windsor P, Ouyang H, G da Costa JA, Rama Damodaran A, Chen Y, Bhagi-Damodaran A. Gas Tunnel Engineering of Prolyl Hydroxylase Reprograms Hypoxia Signaling in Cells. Angew Chem Int Ed Engl 2024; 63:e202409234. [PMID: 39168829 DOI: 10.1002/anie.202409234] [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: 05/15/2024] [Revised: 08/02/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024]
Abstract
Cells have evolved intricate mechanisms for recognizing and responding to changes in oxygen (O2) concentrations. Here, we have reprogrammed cellular hypoxia (low O2) signaling via gas tunnel engineering of prolyl hydroxylase 2 (PHD2), a non-heme iron dependent O2 sensor. Using computational modeling and protein engineering techniques, we identify a gas tunnel and critical residues therein that limit the flow of O2 to PHD2's catalytic core. We show that systematic modification of these residues can open the constriction topology of PHD2's gas tunnel. Using kinetic stopped-flow measurements with NO as a surrogate diatomic gas, we demonstrate up to 3.5-fold enhancement in its association rate to the iron center of tunnel-engineered mutants. Our most effectively designed mutant displays 9-fold enhanced catalytic efficiency (kcat/KM=830±40 M-1 s-1) in hydroxylating a peptide mimic of hypoxia inducible transcription factor HIF-1α, as compared to WT PHD2 (kcat/KM=90±9 M-1 s-1). Furthermore, transfection of plasmids that express designed PHD2 mutants in HEK-293T mammalian cells reveal significant reduction of HIF-1α and downstream hypoxia response transcripts under hypoxic conditions of 1 % O2. Overall, these studies highlight activation of PHD2 as a new pathway to reprogram hypoxia responses and HIF signaling in cells.
Collapse
Affiliation(s)
- Peter Windsor
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Haiping Ouyang
- Department of Biochemistry and Molecular Biology, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Joseph A G da Costa
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Anoop Rama Damodaran
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Yue Chen
- Department of Biochemistry and Molecular Biology, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Ambika Bhagi-Damodaran
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| |
Collapse
|
30
|
Lin TK, Huang CR, Lin KJ, Hsieh YH, Chen SD, Lin YC, Chao AC, Yang DI. Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer's Disease: Beneficial or Detrimental? Antioxidants (Basel) 2024; 13:1378. [PMID: 39594520 PMCID: PMC11591038 DOI: 10.3390/antiox13111378] [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: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The major pathological characteristics of Alzheimer's disease (AD) include senile plaques and neurofibrillary tangles (NFTs), which are mainly composed of aggregated amyloid-beta (Aβ) peptide and hyperphosphorylated tau protein, respectively. The excessive production of reactive oxygen species (ROS) and neuroinflammation are crucial contributing factors to the pathological mechanisms of AD. Hypoxia-inducible factor-1 (HIF-1) is a transcription factor critical for tissue adaption to low-oxygen tension. Growing evidence has suggested HIF-1 as a potential therapeutic target for AD; conversely, other experimental findings indicate that HIF-1 induction contributes to AD pathogenesis. These previous findings thus point to the complex, even contradictory, roles of HIF-1 in AD. In this review, we first introduce the general pathogenic mechanisms of AD as well as the potential pathophysiological roles of HIF-1 in cancer, immunity, and oxidative stress. Based on current experimental evidence in the literature, we then discuss the possible beneficial as well as detrimental mechanisms of HIF-1 in AD; these sections also include the summaries of multiple chemical reagents and proteins that have been shown to exert beneficial effects in AD via either the induction or inhibition of HIF-1.
Collapse
Affiliation(s)
- Tsu-Kung Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Chi-Ren Huang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Kai-Jung Lin
- Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
| | - Yi-Heng Hsieh
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
| | - Yi-Chun Lin
- Department of Neurology, Taipei City Hospital Renai Branch, Taipei 106243, Taiwan;
| | - A-Ching Chao
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Department of Neurology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Ding-I Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
31
|
Yang S, Lin M, Hao S, Ye H, Zhang X. Current hotspots and trends in cancer metabolic reprogramming: a scientometric analysis. Front Immunol 2024; 15:1497461. [PMID: 39588377 PMCID: PMC11586341 DOI: 10.3389/fimmu.2024.1497461] [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: 09/17/2024] [Accepted: 10/18/2024] [Indexed: 11/27/2024] Open
Abstract
Background Metabolic reprogramming (MR) in cancer (CA) has been a focus of intense research in the recent two decades. This phenomenon has attracted great interest because it offers potential targets for cancer therapy. To capture the intellectual landscape of this field, we conducted a bibliometric analysis to assess the scientific output, major contributors, and trends in the MR/CA research. Methods We performed a systematic search using the Web of Science to retrieve articles published on MR of cancer from 2006 until 2023. The bibliometric tools such as Biblioshiny, VOSviewer, and Microsoft Excel were used to identify the most prolific authors, institutions, citation patterns, and keywords. We also used co-citation analysis to map the conceptual structure of the field and identify influential publications. Furthermore, we examined the literature by analyzing publication years, citations, and research impact factors. Results A total of 4,465 publications about MR/CA were retrieved. Publications on MR/CA increased rapidly from 2006 to 2023. Frontiers in Oncology published the most papers, while Cell Metabolism had the most citations. Highly cited papers were mainly published in Cancer Cell, Nature, Cell, Science and Cell Metabolism. China and the United States led the way in publications and contributed the most to MR/CA research. The University of Texas System, Chinese Academy of Sciences, and Fudan University were the most productive institutions. The profitable authors were Deberardinis Ralph J and Chiarugi Paola. The current topics included MR in tumorigenesis and progression of CA, MR of tumor cells and tumor microenvironment, the effect of MR on the CA treatment, the underlying mechanisms of MR (such as gene regulation, epigenetics, extracellular vesicles, and gut microbiota), and the modulation of MR. Some topics such as tumor microenvironment, lipid MR, circular RNA, long noncoding RNA, exosome, prognostic model, and immunotherapy may be the focus of MR/CA research in the next few years. Conclusion This study evaluated the global scientific output in the field of MR/CA research, analyzing its quantitative characteristics. It identified some significant and distinguished papers and compiled information regarding the current status and evolving trends of MR/CA research.
Collapse
Affiliation(s)
- Shanshan Yang
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| | - Miaomiao Lin
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| | - Shaodong Hao
- Spleen and Stomach Disease Department, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Ye
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| | - Xuezhi Zhang
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| |
Collapse
|
32
|
Choya-Foces C, Navarro E, Ríos CDL, López MG, Egea J, Hernansanz-Agustín P, Martínez-Ruiz A. The mitochondrial Na +/Ca 2+ exchanger NCLX is implied in the activation of hypoxia-inducible factors. Redox Biol 2024; 77:103364. [PMID: 39341036 PMCID: PMC11470253 DOI: 10.1016/j.redox.2024.103364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Eukaryotic cells and organisms depend on oxygen for basic living functions, and they display a panoply of adaptations to situations in which oxygen availability is diminished (hypoxia). A number of these responses in animals are mediated by changes in gene expression programs directed by hypoxia-inducible factors (HIFs), whose main mechanism of stabilization and functional activation in response to decreased cytosolic oxygen concentration was elucidated two decades ago. Human acute responses to hypoxia have been known for decades, although their precise molecular mechanism for oxygen sensing is not fully understood. It is already known that a redox component, linked with reactive oxygen species (ROS) production of mitochondrial origin, is implied in these responses. We have recently described a mechanism by which the mitochondrial sodium/calcium exchanger, NCLX, participates in mitochondrial electron transport chain regulation and ROS production in response to acute hypoxia. Here we show that NCLX is also implied in the response to hypoxia mediated by the HIFs. By using a NCLX inhibitor and interference RNA we show that NCLX activity is necessary for HIF-α subunits stabilization in hypoxia and for HIF-1-dependent transcriptional activity. We also show that hypoxic mitochondrial ROS production is not required for HIF-1α stabilization under all circumstances, suggesting that the basal cytosolic redox state or other mechanism(s) could be operating in the NCLX-mediated response to hypoxia that operates through HIF-α stabilization. This finding provides a link between acute and medium-term responses to hypoxia, reinforcing a central role of mitochondrial cell signalling in the response to hypoxia.
Collapse
Affiliation(s)
- Carmen Choya-Foces
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Elisa Navarro
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Cristóbal de Los Ríos
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Grupo de Investigación de Alto Rendimiento en Fisiopatología y Farmacología del Sistema Digestivo (NeuGut), Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón (Madrid), Spain
| | - Manuela G López
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Javier Egea
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Pablo Hernansanz-Agustín
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
| | - Antonio Martínez-Ruiz
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain.
| |
Collapse
|
33
|
Ma Y, Wang Y, Tuo P, Meng Z, Jiang B, Yuan Y, Ding Y, Naeem A, Guo X, Wang X. Downregulation of C1R promotes hepatocellular carcinoma development by activating HIF-1α-regulated glycolysis. Mol Carcinog 2024; 63:2237-2253. [PMID: 39150096 DOI: 10.1002/mc.23806] [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: 03/12/2024] [Revised: 07/11/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024]
Abstract
C1R has been identified to have a distinct function in cutaneous squamous cell carcinoma that goes beyond its role in the complement system. However, it is currently unknown whether C1R is involved in the progression of hepatocellular carcinoma (HCC). HCC tissues were used to examine C1R expression in relation to clinical and pathological factors. Malignant characteristics of HCC cells were assessed through in vitro and in vivo experiments. The mechanism underlying the role of C1R in HCC was explored through RNA-seq, methylation-specific PCR, immuno-precipitation, and dual-luciferase reporter assays. This study found that the expression of C1R decreased as the malignancy of HCC increased and was associated with poor prognosis. C1R promoter was highly methylated through DNMT1 and DNMT3a, resulting in a decrease in C1R expression. Downregulation of C1R expression resulted in heightened malignant characteristics of HCC cells through the activation of HIF-1α-mediated glycolysis. Additionally, decreased C1R expression was found to promote xenograft tumor formation. We found that C-reactive protein (CRP) binds to C1R, and the free CRP activates the NF-κB signaling pathway, which in turn boosts the expression of HIF-1α. This increase in HIF-1α leads to higher glycolysis levels, ultimately promoting aggressive behavior in HCC. Methylation of the C1R promoter region results in the downregulation of C1R expression in HCC. C1R inhibits aggressive behavior in HCC in vitro and in vivo by inhibiting HIF-1α-regulated glycolysis. These findings indicate that C1R acts as a tumor suppressor gene during HCC progression, opening up new possibilities for innovative therapeutic approaches.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Glycolysis/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Animals
- Gene Expression Regulation, Neoplastic
- Mice
- Down-Regulation
- DNA Methylation
- Promoter Regions, Genetic
- Male
- Cell Line, Tumor
- Mice, Nude
- Female
- Prognosis
- Cell Proliferation
- C-Reactive Protein/genetics
- C-Reactive Protein/metabolism
- Signal Transduction
- DNA Methyltransferase 3A/metabolism
- DNA Methyltransferase 3A/genetics
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Yuying Ma
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yuehua Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Peng Tuo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Zhongji Meng
- Department of Infectious Diseases, Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Shiyan, China
| | - Bin Jiang
- Department of Hepatobiliary Pancreatic Surgery, Taihe Hospital, Shiyan, China
| | - Yahong Yuan
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yan Ding
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Abid Naeem
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Xingrong Guo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaoli Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
34
|
Chen C, Xie Y, Qian S. Multifaceted role of GCN2 in tumor adaptation and therapeutic targeting. Transl Oncol 2024; 49:102096. [PMID: 39178574 PMCID: PMC11388189 DOI: 10.1016/j.tranon.2024.102096] [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] [Revised: 07/26/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024] Open
Abstract
Tumor cells voraciously consume nutrients from their environment to facilitate rapid proliferation, necessitating effective strategies to manage nutrient scarcity during tumor growth and progression. A pivotal regulatory mechanism in this context is the Integrated Stress Response (ISR), which ensures cellular homeostasis under conditions such as endoplasmic reticulum stress, the unfolded protein response, and nutrient deprivation. Within the ISR framework, the kinase GCN2 is critical, orchestrating a myriad of cellular processes including the inhibition of protein synthesis, the enhancement of amino acid transport, autophagy initiation, and angiogenesis. These processes collectively enable tumor survival and adaptation under nutrient-limited conditions. Furthermore, GCN2-mediated pathways may induce apoptosis, a property exploited by specific therapeutic agents. Leveraging extensive datasets from TCGA, GEO, and GTEx projects, we conducted a pan-cancer analysis to investigate the prognostic significance of GCN2 expression across diverse cancer types. Our analysis indicates that GCN2 expression significantly varies and correlates with both adverse and favorable prognoses depending on the type of cancer, illustrating its complex role in tumorigenesis. Importantly, GCN2 also modulates the tumor immune microenvironment, influencing immune checkpoint expression and the functionality of immune cells, thereby affecting immunotherapy outcomes. This study highlights the potential of targeting GCN2 with specific inhibitors, as evidenced by their efficacy in preclinical models to augment treatment responses and combat resistance in oncology. These findings advocate for a deeper exploration of GCN2's multifaceted roles, which could pave the way for novel targeted therapies in cancer treatment, aiming to improve clinical outcomes.
Collapse
Affiliation(s)
- Can Chen
- Department of Hematology, Affiliated Hangzhou First People's Hospital, Westlake University, School of Medicine, Hangzhou, China; Zhejiang University, School of Medicine, Hangzhou, China
| | - Yaping Xie
- Department of Hematology, Affiliated Hangzhou First People's Hospital, Westlake University, School of Medicine, Hangzhou, China; Zhejiang University, School of Medicine, Hangzhou, China.
| | - Shenxian Qian
- Department of Hematology, Affiliated Hangzhou First People's Hospital, Westlake University, School of Medicine, Hangzhou, China; Zhejiang University, School of Medicine, Hangzhou, China.
| |
Collapse
|
35
|
Yan P, Yang K, Xu M, Zhu M, Duan Y, Li W, Liu L, Liang C, Li Z, Pan X, Wang L, Yu G. CCT6A alleviates pulmonary fibrosis by inhibiting HIF-1α-mediated lactate production. J Mol Cell Biol 2024; 16:mjae021. [PMID: 38760881 PMCID: PMC11574388 DOI: 10.1093/jmcb/mjae021] [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: 11/15/2023] [Revised: 03/03/2024] [Accepted: 05/16/2024] [Indexed: 05/20/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal progressive fibrotic lung disease. The development of IPF involves different molecular and cellular processes, and recent studies indicate that lactate plays a significant role in promoting the progression of the disease. Nevertheless, the mechanism by which lactate metabolism is regulated and the downstream effects remain unclear. The molecular chaperone CCT6A performs multiple functions in a variety of biological processes. Our research has identified a potential association between CCT6A and serum lactate levels in IPF patients. Herein, we found that CCT6A was highly expressed in type 2 alveolar epithelial cells (AEC2s) of fibrotic lung tissues and correlated with disease severity. Lactate increases the accumulation of lipid droplets in epithelial cells. CCT6A inhibits lipid synthesis by blocking the production of lactate in AEC2s and alleviates bleomycin-induced pulmonary fibrosis in mice. In addition, our results revealed that CCT6A blocks HIF-1α-mediated lactate production by driving the VHL-dependent ubiquitination and degradation of HIF-1α and further inhibits lipid accumulation in fibrotic lungs. In conclusion, we propose that there is a pivotal regulatory role of CCT6A in lactate metabolism in pulmonary fibrosis, and strategies aimed at targeting these key molecules could represent potential therapeutic approaches for pulmonary fibrosis.
Collapse
Affiliation(s)
- Peishuo Yan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Kun Yang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Mengwei Xu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Miaomiao Zhu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Yudi Duan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Wenwen Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Lulu Liu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Chenxi Liang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Zhongzheng Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Xin Pan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
36
|
Fu L, Huang Y, Shan X, Sun X, Wang X, Wang X, Chen L, Yu S. NIR-activatable nitric oxide generator based on nanoparticles loaded small-molecule photosensitizers for synergetic photodynamic/gas therapy. J Nanobiotechnology 2024; 22:595. [PMID: 39354476 PMCID: PMC11446090 DOI: 10.1186/s12951-024-02878-7] [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: 06/12/2024] [Accepted: 09/26/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Therapeutic approaches that combine conventional photodynamic therapy (PDT) with gas therapy (GT) to sensitize PDT are an attractive strategy, but the molecular structure design of the complex lacks effective guiding strategies. RESULTS Herein, we have developed a nanoplatforms Cy-NMNO@SiO2 based on mesoporous silica materials loaded NIR-activatable small-molecule fluorescent probe Cy-NMNO for the synergistic treatment of photodynamic therapy/gas therapy (PDT/GT) in antibacterial and skin cancer. The theoretical calculation results showed that the low dissociation of N-NO in Cy-NMNO enabled it to dissociate effectively under NIR light irradiation, which is conducive to produce Cy and NO. Cy showed better 1O2 generation performance than Cy-NMNO. The cytotoxicity of Cy-NMNO obtained via the synergistic effect of GT and PDT synergistically enhances the effect of photodynamic therapy, thus achieving more effective tumor treatment and sterilization than conventional PDT. Moreover, the nanoplatforms Cy-NMNO@SiO2 realized efficient drug loading and drug delivery. CONCLUSIONS This work not only offers a promising approach for PDT-GT synergistic drug delivery system, but also provides a valuable reference for the design of its drug molecules.
Collapse
Affiliation(s)
- Lili Fu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, Shandong, 250014, China
| | - Yan Huang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Xin Shan
- Department of Intelligent Manufacturing, Shandong City Service Institute, Yantai, 264003, China
| | - Xiao Sun
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Xinlei Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Xiaoyan Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Lingxin Chen
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, China.
| | - Shui Yu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
37
|
Ma QY, Xu XY, Zhu YZ, Yao NN, Liu YC, Gao XD, Zhang Q, Luo WJ. Artesunate inhibits vasculogenic mimicry in choroidal melanoma through HIF-1 α/ VEGF/PDGF pathway. Acta Histochem 2024; 126:152174. [PMID: 38976933 DOI: 10.1016/j.acthis.2024.152174] [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: 01/07/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024]
Abstract
Choroidal melanoma (CM), a highly metastatic eye tumor, exhibits vasculogenic mimicry (VM) facilitated by hypoxia-induced angiogenesis. This study explored the inhibitory impact of the anti-malarial drug Artesunate (ART) on CM VM through modulation of the HIF-1α/VEGF/PDGF pathway. Immunohistochemistry (IHC) confirmed VM in CM with elevated VEGF and PDGF expression. Hypoxia promoted CM proliferation, upregulating HIF-1α, VEGF and PDGF. VEGF and PDGF enhanced CM migration, invasion and VM, with HIF-1α playing a crucial role. ART mitigated VM formation by suppressing the HIF-1α/VEGF/PDGF pathway, highlighting its potential as an anti-tumor agent in CM.
Collapse
Affiliation(s)
- Qing-Yue Ma
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiao-Yan Xu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuan-Zhang Zhu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ning-Ning Yao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yi-Chong Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiao-di Gao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Zhang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen-Juan Luo
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
38
|
Deng Z, Qing Q, Huang B. A bibliometric analysis of the application of the PI3K-AKT-mTOR signaling pathway in cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7255-7272. [PMID: 38709265 DOI: 10.1007/s00210-024-03112-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
PI3K-AKT-mTOR plays as important role in the growth, metabolism, proliferation, and migration of cancer cells, and in apoptosis, autophagy, inflammation, and angiogenesis in cancer. In this study, the aim was to comprehensively review the current research landscape regarding the PI3K-AKT-mTOR pathway in cancer, using bibliometrics to analyze research hotspots, and provide ideas for future research directions. Literature published on the topic between January 2006 and May 2023 was retrieved from the Web of Science core database, and key information and a visualization map were analyzed using CiteSpace and VOSviewer. A total of 5800 articles from 95 countries/regions were collected, including from China and the USA. The number of publications on the topic increased year on year. The major research institution was the University of Texas MD Anderson Cancer Center. Oncotarget and Clinical Cancer Research were the most prevalent journals in the field. Of 26,621 authors, R Kurzrock published the most articles, and J Engelman was cited most frequently. "A549 cell," "first line treatment," "first in human phase I," and "inhibitor" were the keywords of emerging research hotspots. Inhibitors of the PI3K-AKT-mTOR pathway and their use in clinical therapeutic strategies for cancer were the main topics in the field, and future research should also focus on PI3K-AKT-mTOR pathway inhibitors. This study is the first to comprehensively summarize trends and development s in research into the PI3K-AKT-mTOR pathway in cancer. The information that was obtained clarified recent research frontiers and directions, providing references for scholars of cancer management.
Collapse
Affiliation(s)
- Zhengzheng Deng
- School of Public Health, University of South China, Hengyang, 421001, Hunan Province, China
| | - Qiancheng Qing
- School of Public Health, University of South China, Hengyang, 421001, Hunan Province, China
| | - Bo Huang
- School of Public Health, University of South China, Hengyang, 421001, Hunan Province, China.
| |
Collapse
|
39
|
Zhang H, Liu X, Li J, Meng J, Huang W, Su X, Zhang X, Gao G, Wang X, Su H, Zhang F, Zhang T. ING5 inhibits aerobic glycolysis of lung cancer cells by promoting TIE1-mediated phosphorylation of pyruvate dehydrogenase kinase 1 at Y163. Front Med 2024; 18:878-895. [PMID: 39269568 DOI: 10.1007/s11684-024-1057-7] [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: 07/06/2023] [Accepted: 12/04/2023] [Indexed: 09/15/2024]
Abstract
Aerobic glycolysis is critical for tumor growth and metastasis. Previously, we have found that the overexpression of the inhibitor of growth 5 (ING5) inhibits lung cancer aggressiveness and epithelial-mesenchymal transition (EMT). However, whether ING5 regulates lung cancer metabolism reprogramming remains unknown. Here, by quantitative proteomics, we showed that ING5 differentially regulates protein phosphorylation and identified a new site (Y163) of the key glycolytic enzyme PDK1 whose phosphorylation was upregulated 13.847-fold. By clinical study, decreased p-PDK1Y163 was observed in lung cancer tissues and correlated with poor survival. p-PDK1Y163 represents the negative regulatory mechanism of PDK1 by causing PDHA1 dephosphorylation and activation, leading to switching from glycolysis to oxidative phosphorylation, with increasing oxygen consumption and decreasing lactate production. These effects could be impaired by PDK1Y163F mutation, which also impaired the inhibitory effects of ING5 on cancer cell EMT and invasiveness. Mouse xenograft models confirmed the indispensable role of p-PDK1Y163 in ING5-inhibited tumor growth and metastasis. By siRNA screening, ING5-upregulated TIE1 was identified as the upstream tyrosine protein kinase targeting PDK1Y163. TIE1 knockdown induced the dephosphorylation of PDK1Y163 and increased the migration and invasion of lung cancer cells. Collectively, ING5 overexpression-upregulated TIE1 phosphorylates PDK1Y163, which is critical for the inhibition of aerobic glycolysis and invasiveness of lung cancer cells.
Collapse
Affiliation(s)
- Haihua Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xinli Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710038, China
| | - Junqiang Li
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Jin Meng
- Department of Pharmacy, the Medical Security Centre, Chinese PLA General Hospital, Beijing, 100091, China
| | - Wan Huang
- National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi'an, 710038, China
| | - Xuan Su
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xutao Zhang
- Aerospace Clinical Medical Center, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710038, China
| | - Guizhou Gao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xiaodong Wang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| | - Feng Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710038, China.
| | - Tao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
40
|
Spadea A, Tirella A, Rios de la Rosa JM, Lallana E, Mehibel M, Telfer B, Tirelli N, Lawrence MJ, Williams KJ, Stratford IJ, Ashford M. Targeting Hypoxia-Inducible Factor-1α in Pancreatic Cancer: siRNA Delivery Using Hyaluronic Acid-Displaying Nanoparticles. Pharmaceutics 2024; 16:1286. [PMID: 39458615 PMCID: PMC11510765 DOI: 10.3390/pharmaceutics16101286] [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: 08/20/2024] [Revised: 09/16/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Conventional anticancer therapies often lack specificity, targeting both cancerous and normal cells, which reduces efficacy and leads to undesired off-target effects. An additional challenge is the presence of hypoxic regions in tumors, where the Hypoxia Inducible Factor (HIF) transcriptional system drives the expression of pro-survival and drug resistance genes, leading to radio- and chemo-resistance. This study aims to explore the efficacy of targeted nanoparticle (NP)-based small interfering RNA (siRNA) therapies in downregulating these genes to enhance treatment outcomes in pancreatic cancer, a tumor type characterized by high CD44 expression and hypoxia. Methods: We utilized hyaluronic acid (HA)-displaying nanoparticles composed of positively charged chitosan (CS) complexed with siRNA to target and knock down HIF-1α in pancreatic cancer cells. Two NP formulations were prepared using either low molecular weight (LMW) or high molecular weight (HMW) CS. These formulations were evaluated for their internalization by cells and their effectiveness in gene silencing, both in vitro and in vivo. Results: The study found that the molecular weight (MW) of CS influenced the interaction between HA and CD44, as well as the release of siRNA upon internalization. The LMW CS formulation shows faster uptake kinetics, while HMW CS is more effective in gene knockdown across different cell lines in vitro. In vivo, both were able to significantly knockdown HIF-1α and some of its downstream genes. Conclusions: The results suggest that HMW and LMW CS-based NPs exhibit distinct characteristics, showing that both MWs have potential for targeted pancreatic cancer therapy by influencing different aspects of delivery and gene silencing, particularly in the hypoxic tumor microenvironment.
Collapse
Affiliation(s)
- Alice Spadea
- NorthWest Centre for Advanced Drug Delivery (NoWCADD), School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (J.M.R.d.l.R.)
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
| | - Annalisa Tirella
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
- BIOtech Research Centre, Department of Industrial Engineering, University of Trento, 38122 Trento, Italy
| | - Julio Manuel Rios de la Rosa
- NorthWest Centre for Advanced Drug Delivery (NoWCADD), School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (J.M.R.d.l.R.)
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
- Instituto de Investigacion e Innovacion Biomedica de Cadiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cadiz, Spain
| | - Enrique Lallana
- NorthWest Centre for Advanced Drug Delivery (NoWCADD), School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (J.M.R.d.l.R.)
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
- EM Analytical Ltd., Media House, Adlington SK10 4NL, UK
| | - Manal Mehibel
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
- Precision Medicine Oncology, Abbvie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA 94080, USA
| | - Brian Telfer
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
| | - Nicola Tirelli
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
- Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Margaret Jayne Lawrence
- NorthWest Centre for Advanced Drug Delivery (NoWCADD), School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (J.M.R.d.l.R.)
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
| | - Kaye J. Williams
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
| | - Ian J. Stratford
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester M13 9PT, UK; (A.T.); (M.M.); (I.J.S.)
| | - Marianne Ashford
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield M13 9PT, UK;
| |
Collapse
|
41
|
Liu XZ, Tai Y, Hou YB, Cao S, Han J, Li MY, Zuo HX, Xing Y, Jin X, Ma J. Parthenolide Inhibits Synthesis and Promotes Degradation of Programmed Cell Death Ligand 1 and Enhances T Cell Tumor-Killing Activity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21013-21029. [PMID: 39264009 DOI: 10.1021/acs.jafc.4c04916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Parthenolide is a germacrane sesquiterpene lactone separated from the traditional medicinal plant feverfew. Previous studies have shown that parthenolide possesses many pharmacological activities, involving anti-inflammatory and anticancer activities. However, the antitumor mechanism of parthenolide has not been fully elucidated. Thus, we investigate the potential antitumor mechanisms of parthenolactone. We predicted through network pharmacology that parthenolide may target HIF-1α to interfere with the occurrence and development of cancer. We found that parthenolide inhibited PD-L1 protein synthesis through mTOR/p70S6K/4EBP1/eIF4E and RAS/RAF/MEK/MAPK signaling pathways and promoted PD-L1 protein degradation through the lysosomal pathway, thereby inhibiting PD-L1 expression. Immunoprecipitation and Western blotting results demonstrated that parthenolide inhibited PD-L1 expression by suppressing HIF-1α and RAS cooperatively. We further proved that parthenolide inhibited cell proliferation, migration, invasion, and tube formation via down-regulating PD-L1. Moreover, parthenolide increased the effect of T cells to kill tumor cells. In vivo xenograft assays further demonstrated that parthenolide suppressed the growth of tumor xenografts. Collectively, we report for the first time that parthenolide enhanced T cell tumor-killing activity and suppressed cell proliferation, migration, invasion, and tube formation by PD-L1. The current study provides new insight for the development of parthenolide as a novel anticancer drug targeting PD-L1.
Collapse
Affiliation(s)
- Xin Zhe Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Yi Tai
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Yu Bao Hou
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Shen Cao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Jing Han
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Ming Yue Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Hong Xiang Zuo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Yue Xing
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Xuejun Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Juan Ma
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| |
Collapse
|
42
|
Antonelli A, Battaglia AM, Sacco A, Petriaggi L, Giorgio E, Barone S, Biamonte F, Giudice A. Ferroptosis and oral squamous cell carcinoma: connecting the dots to move forward. FRONTIERS IN ORAL HEALTH 2024; 5:1461022. [PMID: 39296524 PMCID: PMC11408306 DOI: 10.3389/froh.2024.1461022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/12/2024] [Indexed: 09/21/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is an aggressive disease whose incomplete biological comprehension contributes to the inappropriate clinical management and poor prognosis. Thus, the identification of new promising molecular targets to treat OSCC is of paramount importance. Ferroptosis is a regulated cell death caused by the iron-dependent accumulation of reactive oxygen species and the consequent oxidative damage of lipid membranes. Over the last five years, a growing number of studies has reported that OSCC is sensitive to ferroptosis induction and that ferroptosis inducers exert a remarkable antitumor effect in OSCC, even in those displaying low response to common approaches, such as chemotherapy and radiotherapy. In addition, as ferroptosis is considered an immunogenic cell death, it may modulate the immune response against OSCC. In this review, we summarize the so far identified ferroptosis regulatory mechanisms and prognostic models based on ferroptosis-related genes in OSCC. In addition, we discuss the perspective of inducing ferroptosis as a novel strategy to directly treat OSCC or, alternatively, to improve sensitivity to other approaches. Finally, we integrate data emerging from the research studies, reviewed here, through in silico analysis and we provide a novel personal perspective on the potential interconnection between ferroptosis and autophagy in OSCC.
Collapse
Affiliation(s)
- Alessandro Antonelli
- Department of Health Science, School of Dentistry, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Anna Martina Battaglia
- Laboratory of Biochemistry and Cellular Biology, Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Alessandro Sacco
- Laboratory of Biochemistry and Cellular Biology, Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Lavinia Petriaggi
- Laboratory of Biochemistry and Cellular Biology, Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Emanuele Giorgio
- Laboratory of Biochemistry and Cellular Biology, Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Selene Barone
- Department of Health Science, School of Dentistry, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Flavia Biamonte
- Laboratory of Biochemistry and Cellular Biology, Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Amerigo Giudice
- Department of Health Science, School of Dentistry, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
43
|
Chen R, Lin Z, Shen S, Zhu C, Yan K, Suo C, Liu R, Wei H, Gao L, Fan K, Zhang H, Sun L, Gao P. Citrullination modulation stabilizes HIF-1α to promote tumour progression. Nat Commun 2024; 15:7654. [PMID: 39227578 PMCID: PMC11372217 DOI: 10.1038/s41467-024-51882-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: 11/21/2023] [Accepted: 08/21/2024] [Indexed: 09/05/2024] Open
Abstract
Citrullination plays an essential role in various physiological or pathological processes, however, whether citrullination is involved in regulating tumour progression and the potential therapeutic significance have not been well explored. Here, we find that peptidyl arginine deiminase 4 (PADI4) directly interacts with and citrullinates hypoxia-inducible factor 1α (HIF-1α) at R698, promoting HIF-1α stabilization. Mechanistically, PADI4-mediated HIF-1αR698 citrullination blocks von Hippel-Lindau (VHL) binding, thereby antagonizing HIF-1α ubiquitination and subsequent proteasome degradation. We also show that citrullinated HIF-1αR698, HIF-1α and PADI4 are highly expressed in hepatocellular carcinoma (HCC) tumour tissues, suggesting a potential correlation between PADI4-mediated HIF-1αR698 citrullination and cancer development. Furthermore, we identify that dihydroergotamine mesylate (DHE) acts as an antagonist of PADI4, which ultimately suppresses tumour progression. Collectively, our results reveal citrullination as a posttranslational modification related to HIF-1α stability, and suggest that targeting PADI4-mediated HIF-1α citrullination is a promising therapeutic strategy for cancers with aberrant HIF-1α expression.
Collapse
Affiliation(s)
- Rui Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Zhiyuan Lin
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Shengqi Shen
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chuxu Zhu
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Kai Yan
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Caixia Suo
- Department of Colorectal Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Rui Liu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Haoran Wei
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Li Gao
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Kaixiang Fan
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Huafeng Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Linchong Sun
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Ping Gao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China.
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
44
|
Mirza Z, Karim S. Unraveling the Mystery of Energy-Sensing Enzymes and Signaling Pathways in Tumorigenesis and Their Potential as Therapeutic Targets for Cancer. Cells 2024; 13:1474. [PMID: 39273044 PMCID: PMC11394487 DOI: 10.3390/cells13171474] [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: 06/06/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 09/15/2024] Open
Abstract
Cancer research has advanced tremendously with the identification of causative genes, proteins, and signaling pathways. Numerous antitumor drugs have been designed and screened for cancer therapeutics; however, designing target-specific drugs for malignant cells with minimal side effects is challenging. Recently, energy-sensing- and homeostasis-associated molecules and signaling pathways playing a role in proliferation, apoptosis, autophagy, and angiogenesis have received increasing attention. Energy-metabolism-based studies have shown the contribution of energetics to cancer development, where tumor cells show increased glycolytic activity and decreased oxidative phosphorylation (the Warburg effect) in order to obtain the required additional energy for rapid division. The role of energy homeostasis in the survival of normal as well as malignant cells is critical; therefore, fuel intake and expenditure must be balanced within acceptable limits. Thus, energy-sensing enzymes detecting the disruption of glycolysis, AMP, ATP, or GTP levels are promising anticancer therapeutic targets. Here, we review the common energy mediators and energy sensors and their metabolic properties, mechanisms, and associated signaling pathways involved in carcinogenesis, and explore the possibility of identifying drugs for inhibiting the energy metabolism of tumor cells. Furthermore, to corroborate our hypothesis, we performed meta-analysis based on transcriptomic profiling to search for energy-associated biomarkers and canonical pathways.
Collapse
Affiliation(s)
- Zeenat Mirza
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21587, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21587, Saudi Arabia;
| | - Sajjad Karim
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21587, Saudi Arabia;
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21587, Saudi Arabia
| |
Collapse
|
45
|
Taneja N, Chauhan A, Kulshreshtha R, Singh S. HIF-1 mediated metabolic reprogramming in cancer: Mechanisms and therapeutic implications. Life Sci 2024; 352:122890. [PMID: 38971364 DOI: 10.1016/j.lfs.2024.122890] [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: 05/06/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Cancer cells undergo metabolic reprogramming to survive in hypoxic conditions and meet the elevated energy demands of the cancer microenvironment. This metabolic alteration is orchestrated by hypoxia-inducible factor 1 (HIF-1), regulating various processes within cancer cells. The intricate metabolic modifications induced by hypoxia underscore the significance of HIF-1-induced metabolic reprogramming in promoting each aspect of cancer progression. The complex interactions between HIF-1 signalling and cellular metabolic processes in response to hypoxia are examined in this study, focusing on the metabolism of carbohydrates, nucleotides, lipids, and amino acids. Comprehending the various regulatory mechanisms controlled by HIF-1 in cellular metabolism sheds light on the intricate biology of cancer growth and offers useful insights for developing targeted treatments.
Collapse
Affiliation(s)
- Nikita Taneja
- Amity Institute of Health Allied Sciences, Amity University, Noida, Uttar Pradesh, India
| | - Akansha Chauhan
- Amity Institute of Health Allied Sciences, Amity University, Noida, Uttar Pradesh, India
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India
| | - Sandhya Singh
- Amity Institute of Health Allied Sciences, Amity University, Noida, Uttar Pradesh, India.
| |
Collapse
|
46
|
Xun Z, Zhou H, Shen M, Liu Y, Sun C, Du Y, Jiang Z, Yang L, Zhang Q, Lin C, Hu Q, Ye Y, Han L. Identification of Hypoxia-ALCAM high Macrophage- Exhausted T Cell Axis in Tumor Microenvironment Remodeling for Immunotherapy Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309885. [PMID: 38956900 PMCID: PMC11434037 DOI: 10.1002/advs.202309885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/02/2024] [Indexed: 07/04/2024]
Abstract
Although hypoxia is known to be associated with immune resistance, the adaptability to hypoxia by different cell populations in the tumor microenvironment and the underlying mechanisms remain elusive. This knowledge gap has hindered the development of therapeutic strategies to overcome tumor immune resistance induced by hypoxia. Here, bulk, single-cell, and spatial transcriptomics are integrated to characterize hypoxia associated with immune escape during carcinogenesis and reveal a hypoxia-based intercellular communication hub consisting of malignant cells, ALCAMhigh macrophages, and exhausted CD8+ T cells around the tumor boundary. A hypoxic microenvironment promotes binding of HIF-1α complex is demonstrated to the ALCAM promoter therefore increasing its expression in macrophages, and the ALCAMhigh macrophages co-localize with exhausted CD8+ T cells in the tumor spatial microenvironment and promote T cell exhaustion. Preclinically, HIF-1ɑ inhibition reduces ALCAM expression in macrophages and exhausted CD8+ T cells and potentiates T cell antitumor function to enhance immunotherapy efficacy. This study reveals the systematic landscape of hypoxia at single-cell resolution and spatial architecture and highlights the effect of hypoxia on immunotherapy resistance through the ALCAMhigh macrophage-exhausted T cell axis, providing a novel immunotherapeutic strategy to overcome hypoxia-induced resistance in cancers.
Collapse
Affiliation(s)
- Zhenzhen Xun
- Center for Immune‐Related Diseases at Shanghai Institute of ImmunologyDepartment of GastroenterologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai Institute of ImmunologyState Key Laboratory of Systems Medicine for CancerDepartment of Immunology and MicrobiologyShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Huanran Zhou
- Department of EndocrinologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Mingyi Shen
- Center for Immune‐Related Diseases at Shanghai Institute of ImmunologyDepartment of GastroenterologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai Institute of ImmunologyState Key Laboratory of Systems Medicine for CancerDepartment of Immunology and MicrobiologyShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Yao Liu
- Department of Hepatobiliary SurgeryCentre for Leading Medicine and Advanced Technologies of IHMThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| | - Chengcao Sun
- Department of Molecular and Cellular OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Yanhua Du
- Center for Immune‐Related Diseases at Shanghai Institute of ImmunologyDepartment of GastroenterologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Zhou Jiang
- Department of Molecular and Cellular OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Liuqing Yang
- Department of Molecular and Cellular OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Qing Zhang
- Simmons Comprehensive Cancer CenterDepartment of PathologyUniversity of Texas Southwestern Medical CenterDallasTX75390USA
| | - Chunru Lin
- Department of Molecular and Cellular OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Qingsong Hu
- Department of Hepatobiliary SurgeryCentre for Leading Medicine and Advanced Technologies of IHMThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| | - Youqiong Ye
- Center for Immune‐Related Diseases at Shanghai Institute of ImmunologyDepartment of GastroenterologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai Institute of ImmunologyState Key Laboratory of Systems Medicine for CancerDepartment of Immunology and MicrobiologyShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Leng Han
- Brown Center for ImmunotherapySchool of MedicineIndiana UniversityIndianapolisIN46202USA
- Department of Biostatistics and Health Data ScienceSchool of MedicineIndiana UniversityIndianapolisIN46202USA
- Department of Biochemistry and Molecular BiologyMcGovern Medical School at The University of Texas Health Science Center at HoustonHoustonTX77030USA
| |
Collapse
|
47
|
Zhang TQ, Lv QY, Jin WL. The cellular-centered view of hypoxia tumor microenvironment: Molecular mechanisms and therapeutic interventions. Biochim Biophys Acta Rev Cancer 2024; 1879:189137. [PMID: 38880161 DOI: 10.1016/j.bbcan.2024.189137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/01/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Cancer is a profoundly dynamic, heterogeneous and aggressive systemic ailment, with a coordinated evolution of various types of tumor niches. Hypoxia plays an indispensable role in the tumor micro-ecosystem, drastically enhancing the plasticity of cancer cells, fibroblasts and immune cells and orchestrating intercellular communication. Hypoxia-induced signals, particularly hypoxia-inducible factor-1α (HIF-1α), drive the reprogramming of genetic, transcriptional, and proteomic profiles. This leads to a spectrum of interconnected processes, including augmented survival of cancer cells, evasion of immune surveillance, metabolic reprogramming, remodeling of the extracellular matrix, and the development of resistance to conventional therapeutic modalities like radiotherapy and chemotherapy. Here, we summarize the latest research on the multifaceted effects of hypoxia, where a multitude of cellular and non-cellular elements crosstalk with each other and co-evolve in a synergistic manner. Additionally, we investigate therapeutic approaches targeting hypoxic niche, encompassing hypoxia-activated prodrugs, HIF inhibitors, nanomedicines, and combination therapies. Finally, we discuss some of the issues to be addressed and highlight the potential of emerging technologies in the treatment of cancer.
Collapse
Affiliation(s)
- Tian-Qi Zhang
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; The Second Hospital of Jilin University, Changchun 130041, China
| | - Qian-Yu Lv
- The Second Hospital of Jilin University, Changchun 130041, China
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
48
|
Zheng JH, Zhu YH, Yang J, Ji PX, Zhao RK, Duan ZH, Yao HF, Jia QY, Yin YF, Hu LP, Li Q, Jiang SH, Huo YM, Liu W, Sun YW, Liu DJ. A CLIC1 network coordinates matrix stiffness and the Warburg effect to promote tumor growth in pancreatic cancer. Cell Rep 2024; 43:114633. [PMID: 39154343 DOI: 10.1016/j.celrep.2024.114633] [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: 01/21/2024] [Revised: 06/19/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) features substantial matrix stiffening and reprogrammed glucose metabolism, particularly the Warburg effect. However, the complex interplay between these traits and their impact on tumor advancement remains inadequately explored. Here, we integrated clinical, cellular, and bioinformatics approaches to explore the connection between matrix stiffness and the Warburg effect in PDAC, identifying CLIC1 as a key mediator. Elevated CLIC1 expression, induced by matrix stiffness through Wnt/β-catenin/TCF4 signaling, signifies poorer prognostic outcomes in PDAC. Functionally, CLIC1 serves as a catalyst for glycolytic metabolism, propelling tumor proliferation. Mechanistically, CLIC1 fortifies HIF1α stability by curbing hydroxylation via reactive oxygen species (ROS). Collectively, PDAC cells elevate CLIC1 levels in a matrix-stiffness-responsive manner, bolstering the Warburg effect to drive tumor growth via ROS/HIF1α signaling. Our insights highlight opportunities for targeted therapies that concurrently address matrix properties and metabolic rewiring, with CLIC1 emerging as a promising intervention point.
Collapse
Affiliation(s)
- Jia-Hao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yu-Heng Zhu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Jian Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Pei-Xuan Ji
- Shanghai Institute of Digestive Disease, Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, P.R. China
| | - Rui-Kang Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Zong-Hao Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hong-Fei Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Qin-Yuan Jia
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yi-Fan Yin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Li-Peng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Qing Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Yan-Miao Huo
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| | - Wei Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| | - Yong-Wei Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| | - De-Jun Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| |
Collapse
|
49
|
Fry LG, Washam CL, Roys H, Bowlin AK, Venugopal G, Bird JT, Byrum SD, Weinkopff T. HIF-α signaling regulates the macrophage inflammatory response during Leishmania major infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.605844. [PMID: 39253467 PMCID: PMC11383058 DOI: 10.1101/2024.08.27.605844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cutaneous leishmaniasis (CL) contributes significantly to the global burden of neglected tropical diseases, with 12 million people currently infected with Leishmania parasites. CL encompasses a range of disease manifestations, from self-healing skin lesions to permanent disfigurations. Currently there is no vaccine available, and many patients are refractory to treatment, emphasizing the need for new therapeutic targets. Previous work demonstrated macrophage HIF-α-mediated lymphangiogenesis is necessary to achieve efficient wound resolution during murine L. major infection. Here, we investigate the role of macrophage HIF-α signaling independent of lymphangiogenesis. We sought to determine the relative contributions of the parasite and the host-mediated inflammation in the lesional microenvironment to myeloid HIF-α signaling. Because HIF-α activation can be detected in infected and bystander macrophages in leishmanial lesions, we hypothesize it is the host's inflammatory response and microenvironment, rather than the parasite, that triggers HIF-α activation. To address this, macrophages from mice with intact HIF-α signaling (LysM Cre ARNT f/+ ) or mice with deleted HIF-α signaling (LysM Cre ARNT f/f ) were subjected to RNASequencing after L. major infection and under pro-inflammatory stimulus. We report that L. major infection alone is enough to induce some minor HIF-α-dependent transcriptomic changes, while infection with L. major in combincation with pro-inflammatory stimuli induces numerous transcriptomic changes that are both dependent and independent of HIF-α signaling. Additionally, by coupling transcriptomic analysis with several pathway analyses, we found HIF-α suppresses pathways involved in protein translation during L. major infection in a pro-inflammatory environment. Together these findings show L. major induces a HIF-α-dependent transcriptomic program, but HIF-α only suppresses protein translation in a pro-inflammatory environment. Thus, this work indicates the host inflammatory response, rather than the parasite, largely contributes to myeloid HIF-α signaling during Leishmania infection.
Collapse
|
50
|
Yang S, Zhan Q, Su D, Cui X, Zhao J, Wang Q, Hong B, Ju J, Cheng C, Yang E, Kang C. HIF1α/ATF3 partake in PGK1 K191/K192 succinylation by modulating P4HA1/succinate signaling in glioblastoma. Neuro Oncol 2024; 26:1405-1420. [PMID: 38441561 PMCID: PMC11300026 DOI: 10.1093/neuonc/noae040] [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] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Hypoxia is a pathological hallmark in most cancers, including glioblastoma (GBM). Hypoxic signaling activation and post-translational modification (PTM) of oncogenic proteins are well-studied in cancers. Accumulating studies indicate glycolytic enzyme PGK1 plays a crucial role in tumorigenesis, yet the underlying mechanisms remain unknown. METHODS We first used ChIP assays to uncover the crosstalk between HIF1α and ATF3 and their roles in P4HA1 regulation. Protein degradation analysis, LC-MS/MS, and in vitro succinate production assays were performed to examine the effect of protein succinylation on GBM pathology. Seahorse assay measured the effects of PGK1 succinylation at K191/K192 or its mutants on glucose metabolism. We utilized an in vivo intracranial mouse model for biochemical studies to elucidate the impact of ATF3 and P4HA1 on aerobic glycolysis and the tumor immune microenvironment. RESULTS We demonstrated that HIF1α and ATF3 positively and negatively regulate the transcription of P4HA1, respectively, leading to an increased succinate production and increased activation of HIF1α signaling. P4HA1 expression elevated the succinate concentration, resulting in the enhanced succinylation of PGK1 at the K191 and K192 sites. Inhibition of proteasomal degradation of PGK1 by succinylation significantly increased aerobic glycolysis to generate lactate. Furthermore, ATF3 overexpression and P4HA1 knockdown reduced succinate and lactate levels in GBM cells, inhibiting immune responses and tumor growth. CONCLUSIONS Together, our study demonstrates that HIF1α/ATF3 participated in P4HA1/succinate signaling, which is the major regulator of succinate biosynthesis and PGK1 succinylation at K191 and K192 sites in GBM. The P4HA1/succinate pathway might be a novel and promising target for aerobic glycolysis in GBM.
Collapse
Affiliation(s)
- Shixue Yang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Qi Zhan
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Dongyuan Su
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Xiaoteng Cui
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Jixing Zhao
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Qixue Wang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Biao Hong
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Jiasheng Ju
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Chunchao Cheng
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Eryan Yang
- Department of Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunsheng Kang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|