1
|
Li Y, Liu X, Dong Y, Zhou Y. Angiogenesis causes and vasculogenic mimicry formation in the context of cancer stem cells. Biochim Biophys Acta Rev Cancer 2025; 1880:189323. [PMID: 40239849 DOI: 10.1016/j.bbcan.2025.189323] [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: 11/14/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
Tumor occurrence, development, invasion, and metastasis are regulated by multiple mechanisms. Among these, angiogenesis promotes tumor progression mainly by supplying tumor tissue and providing channels for tumor metastasis. Cancer stem cells (CSCs) are another important factor affecting tumor progression by involving in tumor initiation and development, while remaining insensitive to conventional antitumor treatments. Among treatment strategies for them, owing to the existence of alternative angiogenic pathways or the risk of damaging normal stem cells, the clinical effect is not ideal. Angiogenesis and CSCs may influence each other in this process. Tumor angiogenesis can support CSC self-renewal by providing a suitable microenvironment, whereas CSCs can regulate tumor neovascularization and mediate drug resistance to anti-angiogenic therapy. This review summarized the role of vascular niche formed by angiogenesis in CSC self-renewal and stemness maintenance, and the function of CSCs in endothelial progenitor cell differentiation and pro-angiogenic factor upregulation. We also elucidated the malignant loop between CSCs and angiogenesis promoting tumor progression. Additionally, we summarized and proposed therapeutic targets, including blocking tumor-derived endothelial differentiation, inhibiting pro-angiogenic factor upregulation, and directly targeting endothelial-like cells comprising CSCs. And we analyzed the feasibility of these strategies to identify more effective methods to improve tumor treatment.
Collapse
Affiliation(s)
- Ying Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaofang Liu
- Department of Anus and Intestine Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yaodong Dong
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Yingying Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
2
|
DePalma T, Rodriguez M, Kollin L, Hughes K, Jones K, Stagner E, Venere M, Skardal A. A Microfluidic Blood Brain Barrier Model to Study the Influence of Glioblastoma Tumor Cells on BBB Function. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411361. [PMID: 40183747 DOI: 10.1002/smll.202411361] [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: 11/26/2024] [Revised: 02/13/2025] [Indexed: 04/05/2025]
Abstract
The blood brain barrier (BBB) plays an essential role in regulating brain function by controlling the transport of nutrients and preventing toxins from moving from the rest of the body's circulation into the brain. Because it is more selective than most other endothelial barriers, many therapeutic candidates fail to cross the BBB, making it difficult to design novel drugs to treat many pathologies in the brain. In addition, BBB dysfunction is observed in many brain diseases including glioblastoma (GB), an aggressive, universally fatal primary brain tumor. Here, a novel 3D microfluidic model of the BBB is designed using human cells and a brain-mimetic hydrogel. The in vitro BBB model replicates several key functions of the human BBB. This system has low permeability to small molecules and responds to inflammatory cues. The addition of GB cells to the model reveals that BBB function changes in a tumor-cell-population-dependent manner. Some GB cell populations lead to increased diffusive permeability while others induce increased immune cell binding. Together, these results indicate that this model can be used to investigate disease progression and drug delivery in GB.
Collapse
Affiliation(s)
- Thomas DePalma
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Marco Rodriguez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Luke Kollin
- Department of Radiation Oncology, Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Kennedy Hughes
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Katie Jones
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Emerie Stagner
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Monica Venere
- Department of Radiation Oncology, Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
3
|
Liu X, Zhang J, Yi T, Li H, Tang X, Liu D, Wu D, Li Y. Decoding tumor angiogenesis: pathways, mechanisms, and future directions in anti-cancer strategies. Biomark Res 2025; 13:62. [PMID: 40251641 PMCID: PMC12007322 DOI: 10.1186/s40364-025-00779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/13/2025] [Indexed: 04/20/2025] Open
Abstract
Angiogenesis, a crucial process in tumor growth and metastasis, necessitates targeted therapeutic intervention. This review reviews the latest knowledge of anti-angiogenesis targets in tumors, with emphasis on the molecular mechanisms and signaling pathways that regulate this process. We emphasize the tumor microenvironment's role in angiogenesis, examine endothelial cell metabolic changes, and evaluated potential therapeutic strategies targeting the tumor vascular system. At the same time, we analyzed the signaling pathway and molecular mechanism of tumor angiogenesis in detail. In addition, this paper also looks at the development trend of tumor anti-angiogenesis drugs, including their future development direction and challenges, aiming to provide prospective insight into the development of this field. Despite their potential, anti-angiogenic therapies encounter challenges like drug resistance and side effects, necessitating ongoing research to enhance cancer treatment strategies and the efficacy of these therapies.
Collapse
Affiliation(s)
- Xueru Liu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Ting Yi
- Department of Trauma Center, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Dan Liu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Daichao Wu
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China.
| |
Collapse
|
4
|
Liu Y, Wu Z, Li Y, Chen Y, Zhao X, Wu M, Xia Y. Metabolic reprogramming and interventions in angiogenesis. J Adv Res 2025; 70:323-338. [PMID: 38704087 PMCID: PMC11976431 DOI: 10.1016/j.jare.2024.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Endothelial cell (EC) metabolism plays a crucial role in the process of angiogenesis. Intrinsic metabolic events such as glycolysis, fatty acid oxidation, and glutamine metabolism, support secure vascular migration and proliferation, energy and biomass production, as well as redox homeostasis maintenance during vessel formation. Nevertheless, perturbation of EC metabolism instigates vascular dysregulation-associated diseases, especially cancer. AIM OF REVIEW In this review, we aim to discuss the metabolic regulation of angiogenesis by EC metabolites and metabolic enzymes, as well as prospect the possible therapeutic opportunities and strategies targeting EC metabolism. KEY SCIENTIFIC CONCEPTS OF REVIEW In this work, we discuss various aspects of EC metabolism considering normal and diseased vasculature. Of relevance, we highlight that the implications of EC metabolism-targeted intervention (chiefly by metabolic enzymes or metabolites) could be harnessed in orchestrating a spectrum of pathological angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Yun Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zifang Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yikun Li
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yating Chen
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xuan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Miaomiao Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Yaoyao Xia
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
5
|
Wu Y, Xie BB, Zhang BL, Zhuang QX, Liu SW, Pan HM. Apatinib regulates the glycolysis of vascular endothelial cells through PI3K/AKT/PFKFB3 pathway in hepatocellular carcinoma. World J Gastroenterol 2025; 31:102848. [PMID: 40124275 PMCID: PMC11924011 DOI: 10.3748/wjg.v31.i11.102848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/17/2025] [Accepted: 02/11/2025] [Indexed: 03/13/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a prevalent and aggressive malignancy in the Chinese population; the severe vascularization by the tumor makes it difficult to cure. The high incidence and poor survival rates of this disease indicate the search for new therapeutic alternatives. Apatinib became a drug of choice because it inhibits tyrosine kinase activity, mainly through an effect on vascular endothelial growth factor receptor-2, thereby preventing tumor angiogenesis. This mechanism of action makes apatinib effective in the treatment of HCC. AIM To investigate the effect of apatinib on the glycolysis of vascular endothelial cells (VECs). METHODS This present study has investigated the effects of HCC cells on VECs, paying particular attention to changes in the glycolytic activity of VECs. The co-culture system established in the present study examined key cellular functions such as extracellular acidification rate and oxygen consumption rate. It also discusses participation of apatinib in the above processes. Core to the findings is the phosphatidylinositol 3-kinase (PI3K)/AKT/6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) signaling pathway, emphasizing the function of phosphorylated AKT and its interaction with PFKFB3, an essential regulator of glycolysis. In the investigation, molecular mechanisms by which such a pathway could influence the above VECs functions of proliferation, migration, and tube formation were underlined through coimmunoprecipitation analysis. Besides, supplementary in vivo experiments on nude mice provided additional biological relevance to the obtained results. RESULTS The glycolytic metabolism in VECs co-cultured with HCC cells is highly active, and the increased glycolysis in these endothelial cells accelerates the malignant transformation of HCC cells. Apatinib has been shown to inhibit this glycolytic activity in the VECs. It also hinders the development, multiplication, and movement of these cells while encouraging their programmed cell death. Moreover, biological analysis revealed that apatinib mainly influences VECs by regulating the PI3K/AKT signaling pathway. Subsequent research indicated that apatinib blocks the PI3K/AKT/PFKEB3 pathway, which in turn reduces glycolysis in these cells. CONCLUSION Apatinib influences the glycolytic pathway in the VECs of HCC a through the PI3K/AKT/PFKFB3 signaling pathway.
Collapse
Affiliation(s)
- Yi Wu
- Division of Cancer Medicine, Sir Run Run Shaw Medical Center, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Bin-Bin Xie
- Division of Cancer Medicine, Sir Run Run Shaw Medical Center, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Bing-Liang Zhang
- Section of Oncology, Ningxia Hui Autonomous Region General Hospital, Ningxia Medical University, Yinchuan 750000, Ningxia Hui Autonomous Region, China
| | - Qing-Xin Zhuang
- Section of Oncology, Ningxia Hui Autonomous Region General Hospital, Ningxia Medical University, Yinchuan 750000, Ningxia Hui Autonomous Region, China
| | - Shi-Wei Liu
- Section of Oncology, Ningxia Hui Autonomous Region General Hospital, Ningxia Medical University, Yinchuan 750000, Ningxia Hui Autonomous Region, China
| | - Hong-Ming Pan
- Division of Cancer Medicine, Sir Run Run Shaw Medical Center, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| |
Collapse
|
6
|
Lin YY, Warren E, Macklin BL, Ramirez L, Gerecht S. Endothelial-pericyte interactions regulate angiogenesis via VEGFR2 signaling during retinal development and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.08.642174. [PMID: 40161680 PMCID: PMC11952325 DOI: 10.1101/2025.03.08.642174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Pericytes stabilize the microvasculature by enhancing endothelial barrier integrity, resulting in functional networks. During retinal development, pericyte recruitment is crucial for stabilizing nascent angiogenic vasculature. However, in adulthood, disrupted endothelial-pericyte interactions lead to vascular dropout and pathological angiogenesis in ocular microvascular diseases, and strategies to stabilize the retinal vasculature are lacking. We demonstrate that direct endothelial-pericyte contact downregulates pVEGFR2 in endothelial cells, which enhances pericyte migration and promotes endothelial cell barrier function. Intravitreal injection of a VEGFR2 inhibitor in mouse models of the developing retina and oxygen-induced retinopathy increased pericyte recruitment and aided vascular stability. The VEGFR2 inhibitor further rescued ischemic retinopathy by enhancing vascularization and tissue growth while reducing vascular permeability. Our findings offer a druggable target to support the growth of functional and mature microvasculature in ocular microvascular diseases and tissue regeneration overall.
Collapse
|
7
|
Liu Y, Wu H, Liang G. Combined Strategies for Nanodrugs Noninvasively Overcoming the Blood-Brain Barrier and Actively Targeting Glioma Lesions. Biomater Res 2025; 29:0133. [PMID: 39911305 PMCID: PMC11794768 DOI: 10.34133/bmr.0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 02/07/2025] Open
Abstract
Drugs for tumor treatment face various challenges, including poor solubility, poor stability, short blood half-life, nontargeting ability, and strong toxic side effects. Fortunately, nanodrug delivery systems provide excellent solution to these problems. However, nanodrugs for glioma treatment also face some key challenges including overcoming the blood-brain barrier (BBB) and, specifically, accumulation in glioma lesions. In this review, we systematically summarize the advantages and disadvantages of combined strategies for nanodrugs noninvasively overcoming BBB and actively targeting glioma lesions to achieve effective glioma therapy. Common noninvasive strategies for nanodrugs overcoming the BBB include bypassing the BBB via the nose-to-brain route, opening the tight junction of the BBB by focused ultrasound with microbubbles, and transendothelial cell transport by intact cell loading, ligand decoration, or cell membrane camouflage of nanodrugs. Actively targeting glioma lesions after overcoming the BBB is another key factor helping nanodrugs accurately treat in situ gliomas. This aim can also be achieved by loading nanodrugs into intact cells and modifying ligand or cell membrane fragments on the surface of nanodrugs. Targeting decorated nanodrugs can guarantee precise glioma killing and avoid side effects on normal brain tissues that contribute to the specific recognition of glioma lesions. Furthermore, the challenges and prospects of nanodrugs in clinical glioma treatment are discussed.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province 471000, China
| | - Haigang Wu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan Province 475004, China
| | - Gaofeng Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province 471000, China
| |
Collapse
|
8
|
Eldakhakhny BM, Ghoneim FM, Soliman MFM, El-Khair SMA, Elsamanoudy AZ, Almoghrabi YM, Mohie PM, Hassan FE, Elfattah AAA. Modulation of placental angiogenesis by metformin in a rat model of gestational diabetes. Histochem Cell Biol 2025; 163:28. [PMID: 39869176 DOI: 10.1007/s00418-025-02355-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2025] [Indexed: 01/28/2025]
Abstract
Gestational diabetes mellitus (GDM) significantly disrupts placental structure and function, leading to complications such as intrauterine growth restriction (IUGR) and preeclampsia. This study aimed to investigate the effects of GDM on placental histology, angiogenesis, and oxidative stress, as well as evaluate metformin's protective role in mitigating these changes. A total of 60 pregnant Sprague-Dawley rats were divided into four groups: control, metformin-treated, GDM, and GDM with metformin. GDM was induced using streptozotocin (STZ) at 40 mg/kg, and metformin was administered at 200 mg/kg from gestational day (GD) 4 to GD17. Blood glucose and insulin levels were assessed, and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) was calculated. Placentae were weighed and subjected to histological, immunohistochemical, and molecular analyses, focusing on key angiogenesis markers (VEGF, VEGFR, CD31, KLF2) and oxidative stress indicators (MDA, eNOS). GDM increased placental weight, angiogenesis (elevated VEGF, VEGFR, CD31), and oxidative stress (elevated MDA, eNOS). Histopathological changes included villous edema, membrane rupture, and hemosiderin deposition. Metformin treatment reduced placental weight; normalized VEGF, KLF2, and PlGF expression; and improved placental architecture. Additionally, oxidative stress was significantly reduced in metformin-treated GDM rats. In conclusion, GDM induces placental abnormalities, promoting excessive angiogenesis and oxidative stress, potentially leading to IUGR and other complications. Metformin showed protective effects by reducing placental overgrowth and restoring vascular and oxidative balance. These findings suggest that metformin may play a therapeutic role in improving placental health in GDM pregnancies, warranting further investigation into its long-term effects on fetal development and maternal health.
Collapse
Affiliation(s)
- Basmah M Eldakhakhny
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, 21465, Jeddah, Saudi Arabia
- Food, Nutrition, and Lifestyle Research Unit, King Fahd for Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatma M Ghoneim
- Physiological Sciences Department, MBBS Program, Fakeeh College for Medical Sciences, 21461, Jeddah, Saudi Arabia
- Medical Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Mona F M Soliman
- Medical Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Salwa M Abo El-Khair
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Ayman Z Elsamanoudy
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, 21465, Jeddah, Saudi Arabia.
- Food, Nutrition, and Lifestyle Research Unit, King Fahd for Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia.
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Yousef M Almoghrabi
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, 21465, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, Regenerative Medicine Unit, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Passant M Mohie
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Fatma E Hassan
- Faculty of Medicine, Medical Physiology Department, Kasr Alainy, Giza, 11562, Egypt
- Department of Physiology, General Medicine Practice Program, Batterjee Medical College, 21442, Jeddah, Saudi Arabia
| | - Amany A Abd Elfattah
- Medical Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
- Department of Basic Medical Sciences, Faculty of Medicine, King Salman International University, South Sinai, Egypt
| |
Collapse
|
9
|
Katano T, Inagaki A, Imura T, Yamana H, Saito R, Endo Kumata Y, Suzuki S, Hagiwara Y, Ohashi K, Watanabe K, Tabata Y, Goto M. A novel approach for hepatocyte transplantation at the liver surface. Cell Transplant 2025; 34:9636897251329308. [PMID: 40208805 PMCID: PMC12032460 DOI: 10.1177/09636897251329308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 04/12/2025] Open
Abstract
Hepatocyte transplantation (HTx) is a promising alternative to liver transplantation; however, poor engraftment remains a major challenge. Although co-transplantation with adipose tissue-derived stromal cells (ADSCs) or islets improves engraftment, exposure of these cells to the portal vein enhances innate immune responses, resulting in a significant loss of hepatocytes. Therefore, we investigated HTx at the liver surface as a novel approach that does not involve the portal vein. Hepatocytes were transplanted onto the liver surface of syngeneic analbuminemic rats with or without ADSCs and/or islets. Serum albumin levels and immunohistochemical staining of the transplanted hepatocytes were evaluated. Hepatocyte engraftment was compared between the liver surface and intraportal groups. To examine the detailed mechanisms behind co-transplantation, co-cultured supernatants were analyzed using multiplex assays, and inhibition tests using neutralizing antibodies were performed. Results showed that islet and ADSC co-transplantation markedly enhanced hepatocyte engraftment at the liver surface (P < 0.01), and its efficiency was comparable to that of intraportal transplantation (P = 0.35). In the co-transplantation group, cells were not necessarily in proximity, suggesting that humoral factors are important. In an in vitro study, hepatocyte function was significantly improved by co-culturing with islets and ADSCs (P < 0.01). Multiplex assays and inhibition tests revealed several important humoral factors, most notably insulin, which promoted hepatocyte engraftment. These findings suggest that HTx at the liver surface, together with crucial factors, may be a novel alternative strategy for intraportal transplantation.
Collapse
Affiliation(s)
- Takumi Katano
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Hiroki Yamana
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Ryusuke Saito
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Yukiko Endo Kumata
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Shoki Suzuki
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Yoshiya Hagiwara
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Kazuo Ohashi
- Graduate School and School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Yasuhiko Tabata
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|
10
|
Ye Y, Cao Z. Glucose Metabolism and Glucose Transporters in Head and Neck Squamous Cell Carcinoma. Cancer Invest 2024; 42:827-844. [PMID: 39324504 DOI: 10.1080/07357907.2024.2407424] [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: 05/11/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Head and neck squamous cell carcinoma ranks seventh globally in malignancy prevalence, with persistent high mortality rates despite treatment advancements. Glucose, pivotal in cancer metabolism via the Warburg effect, enters cells via glucose transporters, notably GLUT proteins. Glycolysis, aerobic oxidation, and the pentose phosphate pathway in glucose metabolism significantly impact HNSCC progression. HNSCC exhibits elevated expression of glucose metabolism enzymes and GLUT proteins, correlating with prognosis. Heterogeneity in HNSCC yields varied metabolic profiles, influenced by factors like HPV status and disease stage. This review highlights glucose metabolism's role and potential as therapeutic targets and cancer imaging tracers in HNSCC.
Collapse
Affiliation(s)
- Yanyan Ye
- Department of Otolaryngology, Shulan (Hangzhou) Hospital, affiliated to Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Zaizai Cao
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Bux K, Asim I, Ismail Z, Hussain S, Herwig R. Structural and dynamical insights revealed the anti-glioblastoma potential of withanolides from Withania coagulans against vascular endothelial growth factor receptor (VEGFR). J Mol Model 2024; 30:383. [PMID: 39443392 DOI: 10.1007/s00894-024-06178-7] [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: 06/29/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
CONTEXT Glioblastoma (GBM), well known as grade 4 tumors due to its progressive malignant features such as vascular proliferation and necrosis, is the most aggressive form of primary brain tumor found in adults. Mutations and amplifications in the vascular endothelial growth factor receptor (VEGFR) contribute to almost 25% of GBM tumors. And thus, VEGFR has been declared the primary target in glioblastoma therapeutic strategies. However, many studies have been previously reported that include GBM as global therapeutics challenge, but they lack the molecular level insights that could help in understanding the biological function of a therapeutically important protein playing a major role in the disease and design the best strategies to develop the potential drugs. METHODS Therefore, to the best of our knowledge, the present study is the first time of kind, which involves multi-in silico approaches to predict the inhibition potential of withanolides from Withania coagulan against VEGFR. The study is actually based on determining the mode of action of five isolates: withanolide J, withaperuvin, 27-hydroxywithanolide I, coagule E, and coagule E, along with their respective binding energies. Molecular docking simulations revealed primarily four ligands, withanolide J (- 7.33 kJ/mol), 27-withanolide (- 7.01 kJ/mol), ajugine, withaperuvin (- 6.89 kJ/mol), and ajugine E (- 6.39 kJ/mol), to have significant binding potencies against the protein. Ligand binding was found to enhance the confirmational stability of the protein revealed through RMSD analysis, and RMSF assessment revealed the protein residues especially from 900-1000 surrounding the binding of the protein. Structural and dynamics of the protein via dynamics cross-correlation movement (DCCM) and principal component analysis (PCA) in both the unbound form and complexed with most potent ligand, withanolide J, reveal the ligand binding affecting the entire conformational integrity of the protein stabilized by hydrogen bonds and electrostatic attractions. Free energy of binding estimations by means of molecular mechanics Poisson-Boltzmann surface area (MMPBSA) method further revealed the withanolide J to have maximum binding potency of the all ligands. Withanolide J in final was also found to have suitable molecular characterizations to cross the blood-brain barrier (BBB +) and reasonable human intestinal absorption ability determined by ADMET profiling via admetSAR tools.
Collapse
Affiliation(s)
- Khair Bux
- Faculty of Life Sciences, Department of Biosciences, Shaheed Zulfikar Ali Bhutto Institute of Science and Technology (SZABIST) University, Karachi, Pakistan.
| | - Irsa Asim
- Faculty of Life Sciences, Department of Biosciences, Shaheed Zulfikar Ali Bhutto Institute of Science and Technology (SZABIST) University, Karachi, Pakistan
| | - Zainab Ismail
- Faculty of Life Sciences, Department of Biosciences, Shaheed Zulfikar Ali Bhutto Institute of Science and Technology (SZABIST) University, Karachi, Pakistan
| | - Samaha Hussain
- Faculty of Life Sciences, Department of Biosciences, Shaheed Zulfikar Ali Bhutto Institute of Science and Technology (SZABIST) University, Karachi, Pakistan
| | - Ralf Herwig
- Laboratories PD Dr. R. Herwig, 80337, Munich, Germany
- Heimerer-College, 10000, Pristina, Kosovo
| |
Collapse
|
12
|
Lochhead JJ, Ronaldson PT, Davis TP. The role of oxidative stress in blood-brain barrier disruption during ischemic stroke: Antioxidants in clinical trials. Biochem Pharmacol 2024; 228:116186. [PMID: 38561092 PMCID: PMC11410550 DOI: 10.1016/j.bcp.2024.116186] [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: 01/22/2024] [Revised: 03/19/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Ischemic stroke is one of the leading causes of death and disability. Occlusion and reperfusion of cerebral blood vessels (i.e., ischemia/reperfusion (I/R) injury) generates reactive oxygen species (ROS) that contribute to brain cell death and dysfunction of the blood-brain barrier (BBB) via oxidative stress. BBB disruption influences the pathogenesis of ischemic stroke by contributing to cerebral edema, hemorrhagic transformation, and extravasation of circulating neurotoxic proteins. An improved understanding of mechanisms for ROS-associated alterations in BBB function during ischemia/reperfusion (I/R) injury can lead to improved treatment paradigms for ischemic stroke. Unfortunately, progress in developing ROS targeted therapeutics that are effective for stroke treatment has been slow. Here, we review how ROS are produced in response to I/R injury, their effects on BBB integrity (i.e., tight junction protein complexes, transporters), and the utilization of antioxidant treatments in ischemic stroke clinical trials. Overall, knowledge in this area provides a strong translational framework for discovery of novel drugs for stroke and/or improved strategies to mitigate I/R injury in stroke patients.
Collapse
Affiliation(s)
- Jeffrey J Lochhead
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| | - Patrick T Ronaldson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Thomas P Davis
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| |
Collapse
|
13
|
Zhao J, Jin D, Huang M, Ji J, Xu X, Wang F, Zhou L, Bao B, Jiang F, Xu W, Lu X, Xiao M. Glycolysis in the tumor microenvironment: a driver of cancer progression and a promising therapeutic target. Front Cell Dev Biol 2024; 12:1416472. [PMID: 38933335 PMCID: PMC11199735 DOI: 10.3389/fcell.2024.1416472] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Even with sufficient oxygen, tumor cells use glycolysis to obtain the energy and macromolecules they require to multiply, once thought to be a characteristic of tumor cells known as the "Warburg effect". In fact, throughout the process of carcinogenesis, immune cells and stromal cells, two major cellular constituents of the tumor microenvironment (TME), also undergo thorough metabolic reprogramming, which is typified by increased glycolysis. In this review, we provide a full-scale review of the glycolytic remodeling of several types of TME cells and show how these TME cells behave in the acidic milieu created by glucose shortage and lactate accumulation as a result of increased tumor glycolysis. Notably, we provide an overview of putative targets and inhibitors of glycolysis along with the viability of using glycolysis inhibitors in combination with immunotherapy and chemotherapy. Understanding the glycolytic situations in diverse cells within the tumor immunological milieu will aid in the creation of subsequent treatment plans.
Collapse
Affiliation(s)
- Junpeng Zhao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Dandan Jin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Mengxiang Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jie Ji
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xuebing Xu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Fei Wang
- Department of Laboratory Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, Jiangsu, China
| | - Lirong Zhou
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Baijun Bao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Feng Jiang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Weisong Xu
- Department of Gastroenterology, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiaomin Lu
- Department of Oncology Affiliated Haian Hospital of Nantong University, Nantong, Jiangsu, China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Laboratory Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
14
|
Xu S, Liao J, Liu B, Zhang C, Xu X. Aerobic glycolysis of vascular endothelial cells: a novel perspective in cancer therapy. Mol Biol Rep 2024; 51:717. [PMID: 38824197 PMCID: PMC11144152 DOI: 10.1007/s11033-024-09588-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/25/2024] [Indexed: 06/03/2024]
Abstract
Vascular endothelial cells (ECs) are monolayers of cells arranged in the inner walls of blood vessels. Under normal physiological conditions, ECs play an essential role in angiogenesis, homeostasis and immune response. Emerging evidence suggests that abnormalities in EC metabolism, especially aerobic glycolysis, are associated with the initiation and progression of various diseases, including multiple cancers. In this review, we discuss the differences in aerobic glycolysis of vascular ECs under normal and pathological conditions, focusing on the recent research progress of aerobic glycolysis in tumor vascular ECs and potential strategies for cancer therapy.
Collapse
Affiliation(s)
- Shenhao Xu
- Department of urology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiahao Liao
- Department of urology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Bing Liu
- Department of urology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Cheng Zhang
- Department of urology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| | - Xin Xu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China.
| |
Collapse
|
15
|
Zhao J, Ma X, Gao P, Han X, Zhao P, Xie F, Liu M. Advancing glioblastoma treatment by targeting metabolism. Neoplasia 2024; 51:100985. [PMID: 38479191 PMCID: PMC10950892 DOI: 10.1016/j.neo.2024.100985] [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: 11/24/2023] [Accepted: 03/04/2024] [Indexed: 03/24/2024]
Abstract
Alterations in cellular metabolism are important hallmarks of glioblastoma(GBM). Metabolic reprogramming is a critical feature as it meets the higher nutritional demand of tumor cells, including proliferation, growth, and survival. Many genes, proteins, and metabolites associated with GBM metabolism reprogramming have been found to be aberrantly expressed, which may provide potential targets for cancer treatment. Therefore, it is becoming increasingly important to explore the role of internal and external factors in metabolic regulation in order to identify more precise therapeutic targets and diagnostic markers for GBM. In this review, we define the metabolic characteristics of GBM, investigate metabolic specificities such as targetable vulnerabilities and therapeutic resistance, as well as present current efforts to target GBM metabolism to improve the standard of care.
Collapse
Affiliation(s)
- Jinyi Zhao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Xuemei Ma
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Peixian Gao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Xueqi Han
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Pengxiang Zhao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Fei Xie
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Mengyu Liu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China; Beijing Molecular Hydrogen Research Center, Beijing, China.
| |
Collapse
|
16
|
Peng Q, Zeng W. The protective role of endothelial GLUT1 in ischemic stroke. Brain Behav 2024; 14:e3536. [PMID: 38747733 PMCID: PMC11095318 DOI: 10.1002/brb3.3536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
OBJECTIVE To provide thorough insight on the protective role of endothelial glucose transporter 1 (GLUT1) in ischemic stroke. METHODS We comprehensively review the role of endothelial GLUT1 in ischemic stroke by narrating the findings concerning biological characteristics of GLUT1 in brain in depth, summarizing the changes of endothelial GLUT1 expression and activity during ischemic stroke, discussing how GLUT1 achieves its neuroprotective effect via maintaining endothelial function, and identifying some outstanding blind spots in current studies. RESULTS Endothelial GLUT1 maintains persistent high glucose and energy requirements of the brain by transporting glucose through the blood-brain barrier, which preserves endothelial function and is beneficial to stroke prognosis. CONCLUSION This review underscores the potential involvement of GLUT1 trafficking, activity modulation, and degradation, and we look forward to more clinical and animal studies to illuminate these mechanisms.
Collapse
Affiliation(s)
- Qiwei Peng
- Department of Critical Care Medicine, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology)Ministry of EducationWuhanChina
| | - Weiqi Zeng
- Department of NeurologyThe First People's Hospital of FoshanFoshanChina
| |
Collapse
|
17
|
Hu T, Allam M, Cai S, Henderson W, Yueh B, Garipcan A, Ievlev AV, Afkarian M, Beyaz S, Coskun AF. Single-cell spatial metabolomics with cell-type specific protein profiling for tissue systems biology. Nat Commun 2023; 14:8260. [PMID: 38086839 PMCID: PMC10716522 DOI: 10.1038/s41467-023-43917-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Metabolic reprogramming in cancer and immune cells occurs to support their increasing energy needs in biological tissues. Here we propose Single Cell Spatially resolved Metabolic (scSpaMet) framework for joint protein-metabolite profiling of single immune and cancer cells in male human tissues by incorporating untargeted spatial metabolomics and targeted multiplexed protein imaging in a single pipeline. We utilized the scSpaMet to profile cell types and spatial metabolomic maps of 19507, 31156, and 8215 single cells in human lung cancer, tonsil, and endometrium tissues, respectively. The scSpaMet analysis revealed cell type-dependent metabolite profiles and local metabolite competition of neighboring single cells in human tissues. Deep learning-based joint embedding revealed unique metabolite states within cell types. Trajectory inference showed metabolic patterns along cell differentiation paths. Here we show scSpaMet's ability to quantify and visualize the cell-type specific and spatially resolved metabolic-protein mapping as an emerging tool for systems-level understanding of tissue biology.
Collapse
Affiliation(s)
- Thomas Hu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Mayar Allam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Shuangyi Cai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Walter Henderson
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Brian Yueh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Anton V Ievlev
- Oak Ridge National Laboratory, Center for Nanophase Materials Sciences, Oak Ridge, TN, USA
| | - Maryam Afkarian
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, CA, USA
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
18
|
Zheng S, Li H, Li Y, Chen X, Shen J, Chen M, Zhang C, Wu J, Sun Q. The emerging role of glycolysis and immune evasion in gastric cancer. Cancer Cell Int 2023; 23:317. [PMID: 38071310 PMCID: PMC10710727 DOI: 10.1186/s12935-023-03169-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 08/21/2024] Open
Abstract
Gastric cancer (GC) is the fifth most common malignancy and the third leading cause of cancer-related deaths worldwide. Similar to other types of tumors, GC cells undergo metabolic reprogramming and switch to a "predominantly glycolytic" metabolic pattern to promote its survival and metastasis, also known as "the Warburg effect", which is characterized by enhanced glucose uptake and lactate production. A large number of studies have shown that targeting cancer cells to enhanced glycolysis is a promising strategy, that can make cancer cells more susceptible to other conventional treatment methods of treatment, including chemotherapy, radiotherapy and immunotherapy, and so on. Therefore, this review summarizes the metabolic characteristics of glycolysis in GC cells and focuses on how abnormal lactate concentration can lead to immunosuppression through its effects on the differentiation, metabolism, and function of infiltrating immune cells, and how targeting this phenomenon may be a potential strategy to improve the therapeutic efficacy of GC.
Collapse
Affiliation(s)
- Shanshan Zheng
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Huaizhi Li
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Yaqi Li
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Xu Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
| | - Junyu Shen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Menglin Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Cancan Zhang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Jian Wu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China.
| | - Qingmin Sun
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China.
| |
Collapse
|
19
|
Wang Q, Yi J, Liu H, Luo M, Yin G, Huang Z. Iguratimod promotes functional recovery after SCI by repairing endothelial cell tight junctions. Exp Neurol 2023; 368:114503. [PMID: 37572946 DOI: 10.1016/j.expneurol.2023.114503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Destruction of the blood-spinal cord barrier (BSCB) after spinal cord injury (SCI) is an important factor promoting the progression of the injury. This study addressed how to repair the BSCB in order to promote the repair of injured spinal cords. Iguratimod (IGU), an anti-rheumatic drug, has been approved for clinical use. A spinal cord injury mouse model and TNF-α-stimulated bEnd.3 cells were used to investigate the effect and mechanism of IGU on injured BSCB. An intracerebroventricular osmotic pump was used to administer drugs to the SCI mouse model. The results showed that the SCI mice in the treatment group had better recovery of neurological function than the control group. Examination of the tissue revealed better repair of the BSCB in injured spinal cords after medication. According to the results from the cell model, IGU promoted the expression of tight junction proteins and reduced cell permeability. Further research found that IGU repaired the barrier function by regulating glycolysis levels in the injured endothelial cells. In studying the mechanism, IGU was found to regulate HIF-1α expression through the NF-κB pathway, thereby regulating the expression of the glycolytic enzymes related to endothelial injury. In summary, IGU promoted functional recovery in vivo by repairing the BSCB. In vitro, IGU regulated the level of glycolysis in the damaged endothelium through the NF-κB pathway, thereby repairing the tight junctions between the endothelium. Therefore, IGU may become a potential drug for treating spinal cord injury.
Collapse
Affiliation(s)
- Qian Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiang Yi
- Department of Orthopedics, Yancheng Third People's Hospital, Yancheng 224008, Jiangsu, China
| | - Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mingran Luo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Zhenfei Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
20
|
Zhang H, Wang Y, Qu M, Li W, Wu D, Cata JP, Miao C. Neutrophil, neutrophil extracellular traps and endothelial cell dysfunction in sepsis. Clin Transl Med 2023; 13:e1170. [PMID: 36629024 PMCID: PMC9832433 DOI: 10.1002/ctm2.1170] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Sepsis is a persistent systemic inflammatory condition involving multiple organ failures resulting from a dysregulated immune response to infection, and one of the hallmarks of sepsis is endothelial dysfunction. During its progression, neutrophils are the first line of innate immune defence against infection. Aside from traditional mechanisms, such as phagocytosis or the release of inflammatory cytokines, reactive oxygen species and other antibacterial substances, activated neutrophils also release web-like structures composed of tangled decondensed DNA, histone, myeloperoxidase and other granules called neutrophil extracellular traps (NETs), which can efficiently ensnare bacteria in the circulation. In contrast, excessive neutrophil activation and NET release may induce endothelial cells to shift toward a pro-inflammatory and pro-coagulant phenotype. Furthermore, neutrophils and NETs can degrade glycocalyx on the endothelial cell surface and increase endothelium permeability. Consequently, the endothelial barrier collapses, contributing to impaired microcirculatory blood flow, tissue hypoperfusion and life-threatening organ failure in the late phase of sepsis.
Collapse
Affiliation(s)
- Hao Zhang
- Department of AnesthesiologyZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key laboratory of Perioperative Stress and ProtectionShanghaiChina
- Department of AnesthesiologyShanghai Medical CollegeFudan University, Shanghai, China
| | - Yanghanzhao Wang
- Department of AnesthesiologyZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key laboratory of Perioperative Stress and ProtectionShanghaiChina
- Department of AnesthesiologyShanghai Medical CollegeFudan University, Shanghai, China
| | - Mengdi Qu
- Department of AnesthesiologyZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key laboratory of Perioperative Stress and ProtectionShanghaiChina
- Department of AnesthesiologyShanghai Medical CollegeFudan University, Shanghai, China
| | - Wenqian Li
- Department of AnesthesiologyZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key laboratory of Perioperative Stress and ProtectionShanghaiChina
| | - Dan Wu
- Department of AnesthesiologyZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key laboratory of Perioperative Stress and ProtectionShanghaiChina
- Department of AnesthesiologyShanghai Medical CollegeFudan University, Shanghai, China
| | - Juan P. Cata
- Department of Anesthesiology and Perioperative MedicineThe University of Texas‐MD Anderson Cancer CenterHoustonTexasUSA
- Anesthesiology and Surgical Oncology Research GroupHoustonTexasUSA
| | - Changhong Miao
- Department of AnesthesiologyZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key laboratory of Perioperative Stress and ProtectionShanghaiChina
- Department of AnesthesiologyShanghai Medical CollegeFudan University, Shanghai, China
| |
Collapse
|
21
|
Kuhn AR, van Bilsen M. Oncometabolism: A Paradigm for the Metabolic Remodeling of the Failing Heart. Int J Mol Sci 2022; 23:ijms232213902. [PMID: 36430377 PMCID: PMC9699042 DOI: 10.3390/ijms232213902] [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: 10/04/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Heart failure is associated with profound alterations in cardiac intermediary metabolism. One of the prevailing hypotheses is that metabolic remodeling leads to a mismatch between cardiac energy (ATP) production and demand, thereby impairing cardiac function. However, even after decades of research, the relevance of metabolic remodeling in the pathogenesis of heart failure has remained elusive. Here we propose that cardiac metabolic remodeling should be looked upon from more perspectives than the mere production of ATP needed for cardiac contraction and relaxation. Recently, advances in cancer research have revealed that the metabolic rewiring of cancer cells, often coined as oncometabolism, directly impacts cellular phenotype and function. Accordingly, it is well feasible that the rewiring of cardiac cellular metabolism during the development of heart failure serves similar functions. In this review, we reflect on the influence of principal metabolic pathways on cellular phenotype as originally described in cancer cells and discuss their potential relevance for cardiac pathogenesis. We discuss current knowledge of metabolism-driven phenotypical alterations in the different cell types of the heart and evaluate their impact on cardiac pathogenesis and therapy.
Collapse
|
22
|
Dickie BR, Jin T, Wang P, Hinz R, Harris W, Boutin H, Parker GJ, Parkes LM, Matthews JC. Quantitative kinetic modelling and mapping of cerebral glucose transport and metabolism using glucoCESL MRI. J Cereb Blood Flow Metab 2022; 42:2066-2079. [PMID: 35748031 PMCID: PMC9580170 DOI: 10.1177/0271678x221108841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chemical-exchange spin-lock (CESL) MRI can map regional uptake and utilisation of glucose in the brain at high spatial resolution (i.e sub 0.2 mm3 voxels). We propose two quantitative kinetic models to describe glucose-induced changes in tissue R1ρ and apply them to glucoCESL MRI data acquired in tumour-bearing and healthy rats. When assuming glucose transport is saturable, the maximal transport capacity (Tmax) measured in normal tissue was 3.2 ± 0.6 µmol/min/mL, the half saturation constant (Kt) was 8.8 ± 2.2 mM, the metabolic rate of glucose consumption (MRglc) was 0.21 ± 0.13 µmol/min/mL, and the cerebral blood volume (vb) was 0.006 ± 0.005 mL/mL. Values in tumour were: Tmax = 7.1 ± 2.7 µmol/min/mL, Kt = 14 ± 1.7 mM, MRglc = 0.22 ± 0.09 µmol/min/mL, vb = 0.030 ± 0.035 mL/mL. Tmax and Kt were significantly higher in tumour tissue than normal tissue (p = 0.006 and p = 0.011, respectively). When assuming glucose uptake also occurs via free diffusion, the free diffusion rate (kd) was 0.061 ± 0.017 mL/min/mL in normal tissue and 0.12 ± 0.042 mL/min/mL in tumour. These parameter estimates agree well with literature values obtained using other approaches (e.g. NMR spectroscopy).
Collapse
Affiliation(s)
- Ben R Dickie
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Tao Jin
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ping Wang
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rainer Hinz
- Division of Informatics, Imaging, and Data Science, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - William Harris
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Hervé Boutin
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Geoff Jm Parker
- Bioxydyn Limited, Manchester, UK.,Centre for Medical Image Computing, Department of Medical Physics & Biomedical Engineering and Department of Neuroinflammation, University College London, London, UK
| | - Laura M Parkes
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Julian C Matthews
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
23
|
Kugeratski FG, Santi A, Zanivan S. Extracellular vesicles as central regulators of blood vessel function in cancer. Sci Signal 2022; 15:eaaz4742. [PMID: 36166511 DOI: 10.1126/scisignal.aaz4742] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Blood vessels deliver oxygen and nutrients that sustain tumor growth and enable the dissemination of cancer cells to distant sites and the recruitment of intratumoral immune cells. In addition, the structural and functional abnormalities of the tumor vasculature foster the development of an aggressive tumor microenvironment and impair the efficacy of existing cancer therapies. Extracellular vesicles (EVs) have emerged as major players of tumor progression, and a growing body of evidence has demonstrated that EVs derived from cancer cells trigger multiple responses in endothelial cells that alter blood vessel function in tumors. EV-mediated signaling in endothelial cells can occur through the transfer of functional cargos such as miRNAs, lncRNAs, cirRNAs, and proteins. Moreover, membrane-bound proteins in EVs can elicit receptor-mediated signaling in endothelial cells. Together, these mechanisms reprogram endothelial cells and contribute to the sustained exacerbated angiogenic signaling typical of tumors, which, in turn, influences cancer progression. Targeting these angiogenesis-promoting EV-dependent mechanisms may offer additional strategies to normalize tumor vasculature. Here, we discuss the current knowledge pertaining to the contribution of cancer cell-derived EVs in mechanisms regulating blood vessel functions in tumors. Moreover, we discuss the translational opportunities in targeting the dysfunctional tumor vasculature using EVs and highlight the open questions in the field of EV biology that can be addressed using mass spectrometry-based proteomics analysis.
Collapse
Affiliation(s)
- Fernanda G Kugeratski
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Alice Santi
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, 50134 Firenze, Italy
| | - Sara Zanivan
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1QH, UK
| |
Collapse
|
24
|
Metabolic Reprogramming in Tumor Endothelial Cells. Int J Mol Sci 2022; 23:ijms231911052. [PMID: 36232355 PMCID: PMC9570383 DOI: 10.3390/ijms231911052] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/29/2022] Open
Abstract
The dynamic crosstalk between the different components of the tumor microenvironment is critical to determine cancer progression, metastatic dissemination, tumor immunity, and therapeutic responses. Angiogenesis is critical for tumor growth, and abnormal blood vessels contribute to hypoxia and acidosis in the tumor microenvironment. In this hostile environment, cancer and stromal cells have the ability to alter their metabolism in order to support the high energetic demands and favor rapid tumor proliferation. Recent advances have shown that tumor endothelial cell metabolism is reprogrammed, and that targeting endothelial metabolic pathways impacts developmental and pathological vessel sprouting. Therefore, the use of metabolic antiangiogenic therapies to normalize the blood vasculature, in combination with immunotherapies, offers a clinical niche to treat cancer.
Collapse
|
25
|
Shi X, Yang J, Deng S, Xu H, Wu D, Zeng Q, Wang S, Hu T, Wu F, Zhou H. TGF-β signaling in the tumor metabolic microenvironment and targeted therapies. J Hematol Oncol 2022; 15:135. [PMID: 36115986 PMCID: PMC9482317 DOI: 10.1186/s13045-022-01349-6] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/24/2022] [Indexed: 12/30/2022] Open
Abstract
AbstractTransforming growth factor-β (TGF-β) signaling has a paradoxical role in cancer progression, and it acts as a tumor suppressor in the early stages but a tumor promoter in the late stages of cancer. Once cancer cells are generated, TGF-β signaling is responsible for the orchestration of the immunosuppressive tumor microenvironment (TME) and supports cancer growth, invasion, metastasis, recurrence, and therapy resistance. These progressive behaviors are driven by an “engine” of the metabolic reprogramming in cancer. Recent studies have revealed that TGF-β signaling regulates cancer metabolic reprogramming and is a metabolic driver in the tumor metabolic microenvironment (TMME). Intriguingly, TGF-β ligands act as an “endocrine” cytokine and influence host metabolism. Therefore, having insight into the role of TGF-β signaling in the TMME is instrumental for acknowledging its wide range of effects and designing new cancer treatment strategies. Herein, we try to illustrate the concise definition of TMME based on the published literature. Then, we review the metabolic reprogramming in the TMME and elaborate on the contribution of TGF-β to metabolic rewiring at the cellular (intracellular), tissular (intercellular), and organismal (cancer-host) levels. Furthermore, we propose three potential applications of targeting TGF-β-dependent mechanism reprogramming, paving the way for TGF-β-related antitumor therapy from the perspective of metabolism.
Collapse
|
26
|
The role of metabolic reprogramming in cancer metastasis and potential mechanism of traditional Chinese medicine intervention. Biomed Pharmacother 2022; 153:113376. [DOI: 10.1016/j.biopha.2022.113376] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 11/22/2022] Open
|
27
|
Rahman MM, Islam MR, Yamin M, Islam MM, Sarker MT, Meem AFK, Akter A, Emran TB, Cavalu S, Sharma R. Emerging Role of Neuron-Glia in Neurological Disorders: At a Glance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3201644. [PMID: 36046684 PMCID: PMC9423989 DOI: 10.1155/2022/3201644] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022]
Abstract
Based on the diverse physiological influence, the impact of glial cells has become much more evident on neurological illnesses, resulting in the origins of many diseases appearing to be more convoluted than previously happened. Since neurological disorders are often random and unknown, hence the construction of animal models is difficult to build, representing a small fraction of people with a gene mutation. As a result, an immediate necessity is grown to work within in vitro techniques for examining these illnesses. As the scientific community recognizes cell-autonomous contributions to a variety of central nervous system illnesses, therapeutic techniques involving stem cells for treating neurological diseases are gaining traction. The use of stem cells derived from a variety of sources is increasingly being used to replace both neuronal and glial tissue. The brain's energy demands necessitate the reliance of neurons on glial cells in order for it to function properly. Furthermore, glial cells have diverse functions in terms of regulating their own metabolic activities, as well as collaborating with neurons via secreted signaling or guidance molecules, forming a complex network of neuron-glial connections in health and sickness. Emerging data reveals that metabolic changes in glial cells can cause morphological and functional changes in conjunction with neuronal dysfunction under disease situations, highlighting the importance of neuron-glia interactions in the pathophysiology of neurological illnesses. In this context, it is required to improve our understanding of disease mechanisms and create potential novel therapeutics. According to research, synaptic malfunction is one of the features of various mental diseases, and glial cells are acting as key ingredients not only in synapse formation, growth, and plasticity but also in neuroinflammation and synaptic homeostasis which creates critical physiological capacity in the focused sensory system. The goal of this review article is to elaborate state-of-the-art information on a few glial cell types situated in the central nervous system (CNS) and highlight their role in the onset and progression of neurological disorders.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Yamin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Mohaimenul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Taslim Sarker
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Atkia Farzana Khan Meem
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Aklima Akter
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 Uttar Pradesh, India
| |
Collapse
|
28
|
Sharma V, Singh TG, Mannan A. Therapeutic implications of glucose transporters (GLUT) in cerebral ischemia. Neurochem Res 2022; 47:2173-2186. [PMID: 35596882 DOI: 10.1007/s11064-022-03620-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 01/05/2023]
Abstract
Cerebral ischemia is a leading cause of death in the globe, with a large societal cost. Deprivation of blood flow, together with consequent glucose and oxygen shortage, activates a variety of pathways that result in permanent brain damage. As a result, ischemia raises energy demand, which is linked to significant alterations in brain energy metabolism. Even at the low glucose levels reported in plasma during ischemia, glucose transport activity may adjust to assure the supply of glucose to maintain normal cellular function. Glucose transporters in the brain are divided into two groups: sodium-independent glucose transporters (GLUTs) and sodium-dependent glucose cotransporters (SGLTs).This review assess the GLUT structure, expression, regulation, pathobiology of GLUT in cerebral ischemia and regulators of GLUT and it also provides the synopsis of the literature exploring the relationship between GLUT and the various downstream signalling pathways for e.g., AMP-activated protein kinase (AMPK), CREB (cAMP response element-binding protein), Hypoxia-inducible factor 1 (HIF)-1, Phosphatidylinositol 3-kinase (PI3-K), Mitogen-activated protein kinase (MAPK) and adenylate-uridylate-rich elements (AREs). Therefore, the aim of the present review was to elaborate the therapeutic implications of GLUT in the cerebral ischemia.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, 140401, Patiala, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, 140401, Patiala, Punjab, India.
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, 140401, Patiala, Punjab, India
| |
Collapse
|
29
|
Macklin BL, Lin YY, Emmerich K, Wisniewski E, Polster BM, Konstantopoulos K, Mumm JS, Gerecht S. Intrinsic epigenetic control of angiogenesis in induced pluripotent stem cell-derived endothelium regulates vascular regeneration. NPJ Regen Med 2022; 7:28. [PMID: 35551465 PMCID: PMC9098630 DOI: 10.1038/s41536-022-00223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
Human-induced pluripotent stem cell-derived endothelial cells (iECs) provide opportunities to study vascular development and regeneration, develop cardiovascular therapeutics, and engineer model systems for drug screening. The differentiation and characterization of iECs are well established; however, the mechanisms governing their angiogenic phenotype remain unknown. Here, we aimed to determine the angiogenic phenotype of iECs and the regulatory mechanism controlling their regenerative capacity. In a comparative study with HUVECs, we show that iECs increased expression of vascular endothelial growth factor receptor 2 (VEGFR2) mediates their highly angiogenic phenotype via regulation of glycolysis enzymes, filopodia formation, VEGF mediated migration, and robust sprouting. We find that the elevated expression of VEGFR2 is epigenetically regulated via intrinsic acetylation of histone 3 at lysine 27 by histone acetyltransferase P300. Utilizing a zebrafish xenograft model, we demonstrate that the ability of iECs to promote the regeneration of the amputated fin can be modulated by P300 activity. These findings demonstrate how the innate epigenetic status of iECs regulates their phenotype with implications for their therapeutic potential.
Collapse
Affiliation(s)
- Bria L Macklin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ying-Yu Lin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Emily Wisniewski
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA. .,Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
30
|
Zlacká J, Zeman M. Glycolysis under Circadian Control. Int J Mol Sci 2021; 22:ijms222413666. [PMID: 34948470 PMCID: PMC8703893 DOI: 10.3390/ijms222413666] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/07/2021] [Accepted: 12/17/2021] [Indexed: 12/31/2022] Open
Abstract
Glycolysis is considered a main metabolic pathway in highly proliferative cells, including endothelial, epithelial, immune, and cancer cells. Although oxidative phosphorylation (OXPHOS) is more efficient in ATP production per mole of glucose, proliferative cells rely predominantly on aerobic glycolysis, which generates ATP faster compared to OXPHOS and provides anabolic substrates to support cell proliferation and migration. Cellular metabolism, including glucose metabolism, is under strong circadian control. Circadian clocks control a wide array of metabolic processes, including glycolysis, which exhibits a distinct circadian pattern. In this review, we discuss circadian regulations during metabolic reprogramming and key steps of glycolysis in activated, highly proliferative cells. We suggest that the inhibition of metabolic reprogramming in the circadian manner can provide some advantages in the inhibition of oxidative glycolysis and a chronopharmacological approach is a promising way to treat diseases associated with up-regulated glycolysis.
Collapse
|
31
|
Yang Q, Ma Q, Xu J, Liu Z, Mao X, Zhou Y, Cai Y, Da Q, Hong M, Weintraub NL, Fulton DJ, Belin de Chantemèle EJ, Huo Y. Endothelial AMPKα1/PRKAA1 exacerbates inflammation in HFD-fed mice. Br J Pharmacol 2021; 179:1661-1678. [PMID: 34796475 PMCID: PMC9112062 DOI: 10.1111/bph.15742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Excess nutrient-induced endothelial cell inflammation is a hallmark in high fat diet (HFD)-induced metabolic syndrome. Pharmacological activation of protein kinase AMP-activated alpha 1(PRKAA1)/5'-Adenosine monophosphate-activated protein kinase alpha1 (AMPKα1) shows its beneficial effects in many studies of cardiometabolic disorders. However, AMPKα1, as a major cellular sensor of energy and nutrients in endothelial cells, has not been studied for its physiological role in excess nutrient-induced endothelial cell (EC) inflammation. EXPERIMENTAL APPROACH Wild-type and EC-specific Prkaa1 knockout mice were fed with an HFD. Body weight, fat mass composition, glucose and lipid levels were monitored regularly. Insulin sensitivity was analyzed systemically and in major metabolic organs/tissues. Inflammation status in metabolic organs/tissues were examined with quantitative RT-PCR and flow cytometry. Additionally, metabolic status, inflammation severity and signaling in cultured ECs were assayed with multiple approaches at the molecular level. KEY RESULTS EC Prkaa1 deficiency unexpectedly alleviated HFD-induced metabolic syndromes including decreased body weight and fat mass, enhanced glucose clearance and insulin sensitivity, and relieved adipose inflammation and hepatic steatosis. Mechanistically, PRKAA1 knockdown in cultured ECs reduced endothelial glycolysis and fatty acid oxidation, decreased the levels of acetyl-coA, and suppressed transcription of inflammatory molecules mediated by ATP citrate lyase (ACLY) and histone acetyltransferase p300. CONCLUSIONS AND IMPLICATIONS This unexpected pro-inflammatory effect of endothelial AMPKα1/PRKAA1 in metabolic context provides additional insight in AMPKα1/PRKAA1 activities, warranting that in-depth study and thoughtful consideration should be applied when AMPKα1/PRKAA1 is used as a therapeutic target in the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Qiuhua Yang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Qian Ma
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA.,State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University, Shenzhen, China
| | - Jiean Xu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA.,State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University, Shenzhen, China
| | - Zhiping Liu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiaoxiao Mao
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA.,State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University, Shenzhen, China
| | - Yaqi Zhou
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University, Shenzhen, China
| | - Yongfeng Cai
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University, Shenzhen, China
| | - Qingen Da
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Mei Hong
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University, Shenzhen, China
| | - Neal L Weintraub
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David J Fulton
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
32
|
Shin E, Koo JS. Glucose Metabolism and Glucose Transporters in Breast Cancer. Front Cell Dev Biol 2021; 9:728759. [PMID: 34552932 PMCID: PMC8450384 DOI: 10.3389/fcell.2021.728759] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most common malignancy in women worldwide and is associated with high mortality rates despite the continuously advancing treatment strategies. Glucose is essential for cancer cell metabolism owing to the Warburg effect. During the process of glucose metabolism, various glycolytic metabolites, such as serine and glycine metabolites, are produced and other metabolic pathways, such as the pentose phosphate pathway (PPP), are associated with the process. Glucose is transported into the cell by glucose transporters, such as GLUT. Breast cancer shows high expressions of glucose metabolism-related enzymes and GLUT, which are also related to breast cancer prognosis. Triple negative breast cancer (TNBC), which is a high-grade breast cancer, is especially dependent on glucose metabolism. Breast cancer also harbors various stromal cells such as cancer-associated fibroblasts and immune cells as tumor microenvironment, and there exists a metabolic interaction between these stromal cells and breast cancer cells as explained by the reverse Warburg effect. Breast cancer is heterogeneous, and, consequently, its metabolic status is also diverse, which is especially affected by the molecular subtype, progression stage, and metastatic site. In this review, we will focus on glucose metabolism and glucose transporters in breast cancer, and we will additionally discuss their potential applications as cancer imaging tracers and treatment targets.
Collapse
Affiliation(s)
| | - Ja Seung Koo
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
33
|
Yue L, Lu X, Dennery PA, Yao H. Metabolic dysregulation in bronchopulmonary dysplasia: Implications for identification of biomarkers and therapeutic approaches. Redox Biol 2021; 48:102104. [PMID: 34417157 PMCID: PMC8710987 DOI: 10.1016/j.redox.2021.102104] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/03/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in premature infants. Accumulating evidence shows that dysregulated metabolism of glucose, lipids and amino acids are observed in premature infants. Animal and cell studies demonstrate that abnormal metabolism of these substrates results in apoptosis, inflammation, reduced migration, abnormal proliferation or senescence in response to hyperoxic exposure, and that rectifying metabolic dysfunction attenuates neonatal hyperoxia-induced alveolar simplification and vascular dysgenesis in the lung. BPD is often associated with several comorbidities, including pulmonary hypertension and neurodevelopmental abnormalities, which significantly increase the morbidity and mortality of this disease. Here, we discuss recent progress on dysregulated metabolism of glucose, lipids and amino acids in premature infants with BPD and in related in vivo and in vitro models. These findings suggest that metabolic dysregulation may serve as a biomarker of BPD and plays important roles in the pathogenesis of this disease. We also highlight that targeting metabolic pathways could be employed in the prevention and treatment of BPD.
Collapse
Affiliation(s)
- Li Yue
- Department of Orthopedics, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Xuexin Lu
- Department of Pediatrics, Ascension St. John Hospital, Detroit, MI, USA
| | - Phyllis A Dennery
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, USA; Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, USA.
| |
Collapse
|
34
|
Ullah K, Wu R. Hypoxia-Inducible Factor Regulates Endothelial Metabolism in Cardiovascular Disease. Front Physiol 2021; 12:670653. [PMID: 34290616 PMCID: PMC8287728 DOI: 10.3389/fphys.2021.670653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells (ECs) form a physical barrier between the lumens and vascular walls of arteries, veins, capillaries, and lymph vessels; thus, they regulate the extravasation of nutrients and oxygen from the circulation into the perivascular space and participate in mechanisms that maintain cardiovascular homeostasis and promote tissue growth and repair. Notably, their role in tissue repair is facilitated, at least in part, by their dependence on glycolysis for energy production, which enables them to resist hypoxic damage and promote angiogenesis in ischemic regions. ECs are also equipped with a network of oxygen-sensitive molecules that collectively activate the response to hypoxic injury, and the master regulators of the hypoxia response pathway are hypoxia-inducible factors (HIFs). HIFs reinforce the glycolytic dependence of ECs under hypoxic conditions, but whether HIF activity attenuates or exacerbates the progression and severity of cardiovascular dysfunction varies depending on the disease setting. This review summarizes how HIF regulates the metabolic and angiogenic activity of ECs under both normal and hypoxic conditions and in a variety of diseases that are associated with cardiovascular complications.
Collapse
Affiliation(s)
- Karim Ullah
- Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Rongxue Wu
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
35
|
Yang D, Guo P, He T, Powell CA. Role of endothelial cells in tumor microenvironment. Clin Transl Med 2021; 11:e450. [PMID: 34185401 PMCID: PMC8214858 DOI: 10.1002/ctm2.450] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/21/2022] Open
Affiliation(s)
- Dawei Yang
- Department of Pulmonary and Critical Care MedicineZhongshan Hospital Institute for Clinical Science, Shanghai Medical CollegeShanghai Engineering Research Center of AI Technology for Cardiopulmonary DiseasesShanghai Engineer & Technology Research Center of Internet of Things for Respiratory MedicineZhongshan Hospital Fudan UniversityShanghai200032China
- Division of Pulmonary, Critical Care and Sleep MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Tianrui He
- Department of Pulmonary and Critical Care MedicineZhongshan Hospital Institute for Clinical Science, Shanghai Medical CollegeShanghai Engineering Research Center of AI Technology for Cardiopulmonary DiseasesShanghai Engineer & Technology Research Center of Internet of Things for Respiratory MedicineZhongshan Hospital Fudan UniversityShanghai200032China
| | - Charles A. Powell
- Division of Pulmonary, Critical Care and Sleep MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
36
|
Pharmacological inhibition of GLUT1 as a new immunotherapeutic approach after myocardial infarction. Biochem Pharmacol 2021; 190:114597. [PMID: 33965393 DOI: 10.1016/j.bcp.2021.114597] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 11/21/2022]
Abstract
Myocardial infarction (MI) is one of the major contributors to cardiovascular morbidity and mortality. Excess inflammation significantly contributes to cardiac remodeling and heart failure after MI. Accumulating evidence has shown the central role of cellular metabolism in regulating the differentiation and function of cells. Metabolic rewiring is particularly relevant for proinflammatory responses induced by ischemia. Hypoxia reduces mitochondrial oxidative phosphorylation (OXPHOS) and induces increased reliance on glycolysis. Moreover, activation of a proinflammatory transcriptional program is associated with preferential glucose metabolism in leukocytes. An improved understanding of the mechanisms that regulate metabolic adaptations holds the potential to identify new metabolic targets and strategies to reduce ischemic cardiac damage, attenuate excess local inflammation and ultimately prevent the development of heart failure. Among possible drug targets, glucose transporter 1 (GLUT1) gained considerable interest considering its pivotal role in regulating glucose availability in activated leukocytes and the availability of small molecules that selectively inhibit it. Therefore, we summarize current evidence on the role of GLUT1 in leukocytes (focusing on macrophages and T cells) and non-leukocytes, including cardiomyocytes, endothelial cells and fibroblasts regarding ischemic heart disease. Beyond myocardial infarction, we can foresee the role of GLUT1 blockers as a possible pharmacological approach to limit pathogenic inflammation in other conditions driven by excess sterile inflammation.
Collapse
|
37
|
Certo M, Elkafrawy H, Pucino V, Cucchi D, Cheung KC, Mauro C. Endothelial cell and T-cell crosstalk: Targeting metabolism as a therapeutic approach in chronic inflammation. Br J Pharmacol 2021; 178:2041-2059. [PMID: 31999357 PMCID: PMC8246814 DOI: 10.1111/bph.15002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
The role of metabolic reprogramming in the coordination of the immune response has gained increasing consideration in recent years. Indeed, it has become clear that changes in the metabolic status of immune cells can alter their functional properties. During inflammation, T cells need to generate sufficient energy and biomolecules to support growth, proliferation, and effector functions. Therefore, T cells need to rearrange their metabolism to meet these demands. A similar metabolic reprogramming has been described in endothelial cells, which have the ability to interact with and modulate the function of immune cells. In this overview, we will discuss recent insights in the complex crosstalk between endothelial cells and T cells as well as their metabolic reprogramming following activation. We highlight key components of this metabolic switch that can lead to the development of new therapeutics against chronic inflammatory disorders. LINKED ARTICLES: This article is part of a themed issue on Cellular metabolism and diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.10/issuetoc.
Collapse
Affiliation(s)
- Michelangelo Certo
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Hagar Elkafrawy
- Medical Biochemistry and Molecular Biology Department, Faculty of MedicineAlexandria UniversityAlexandriaEgypt
| | - Valentina Pucino
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Danilo Cucchi
- Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Kenneth C.P. Cheung
- School of Life SciencesThe Chinese University of Hong KongHong Kong SARChina
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Institute of Cardiovascular Sciences, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Institute of Metabolism and Systems Research, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
38
|
Cignarella A, Fadini GP, Bolego C, Trevisi L, Boscaro C, Sanga V, Seccia TM, Rosato A, Rossi GP, Barton M. Clinical Efficacy and Safety of Angiogenesis Inhibitors: Sex Differences and Current Challenges. Cardiovasc Res 2021; 118:988-1003. [PMID: 33739385 DOI: 10.1093/cvr/cvab096] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
Vasoactive molecules, such as vascular endothelial growth factor (VEGF) and endothelins, share cytokine-like activities and regulate endothelial cell (EC) growth, migration and inflammation. Some endothelial mediators and their receptors are targets for currently approved angiogenesis inhibitors, drugs that are either monoclonal antibodies raised towards VEGF, or inhibitors of vascular receptor protein kinases and signaling pathways. Pharmacological interference with the protective functions of ECs results in a similar spectrum of adverse effects. Clinically, the most common side effects of VEGF signaling pathway inhibition include an increase in arterial pressure, left ventricular (LV) dysfunction ultimately causing heart failure, and thromboembolic events, including pulmonary embolism, stroke, and myocardial infarction. Sex steroids such as androgens, progestins, and estrogen and their receptors (ERα, ERβ, GPER; PR-A, PR-B; AR) have been identified as important modifiers of angiogenesis, and sex differences have been reported for anti-angiogenic drugs. This review article discusses the current challenges clinicians are facing with regard to angiogenesis inhibitor treatments, including the need to consider sex differences affecting clinical efficacy and safety. We also propose areas for future research taking into account the role of sex hormone receptors and sex chromosomes. Development of new sex-specific drugs with improved target and cell-type selectivity likely will open the way personalized medicine in men and women requiring antiangiogenic therapy and result in reduced adverse effects and improved therapeutic efficacy.
Collapse
Affiliation(s)
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Lucia Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Carlotta Boscaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Viola Sanga
- Department of Medicine, University of Padova, Italy
| | | | - Antonio Rosato
- Venetian Cancer Institute IOV - IRCCS, Padova, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy
| | | | - Matthias Barton
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy.,Molecular Internal Medicine, University of Zürich, Switzerland.,Andreas Grüntzig Foundation, Zürich, Switzerland
| |
Collapse
|
39
|
Grillo E, Corsini M, Ravelli C, Zammataro L, Bacci M, Morandi A, Monti E, Presta M, Mitola S. Expression of activated VEGFR2 by R1051Q mutation alters the energy metabolism of Sk-Mel-31 melanoma cells by increasing glutamine dependence. Cancer Lett 2021; 507:80-88. [PMID: 33744390 DOI: 10.1016/j.canlet.2021.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022]
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) activating mutations are emerging as important oncogenic driver events. Understanding the biological implications of such mutations may help to pinpoint novel therapeutic targets. Here we show that activated VEGFR2 via the pro-oncogenic R1051Q mutation induces relevant metabolic changes in melanoma cells. The expression of VEGFR2R1051Q leads to higher energy metabolism and ATP production compared to control cells expressing VEGFR2WT. Furthermore, activated VEGFR2R1051Q augments the dependence on glutamine (Gln) of melanoma cells, thus increasing Gln uptake and their sensitivity to Gln deprivation and to inhibitors of glutaminase, the enzyme initiating Gln metabolism by cells. Overall, these results highlight Gln addiction as a metabolic vulnerability of tumors harboring the activating VEGFR2R1051Q mutation and suggest novel therapeutic approaches for those patients harboring activating mutations of VEGFR2.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy.
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Luca Zammataro
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Marina Bacci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, 50134, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, 50134, Italy
| | - Eugenio Monti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy.
| |
Collapse
|
40
|
Li H, Wu H, Wang Q, Ning S, Xu S, Pang D. Dual effects of N 6-methyladenosine on cancer progression and immunotherapy. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 24:25-39. [PMID: 33738136 PMCID: PMC7933696 DOI: 10.1016/j.omtn.2021.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
According to the latest global cancer statistics, cancer has become a major threat to human health, but cancer treatment has encountered many bottlenecks. As an emerging topic in epigenetics, N6-methyladenosine (m6A) is the most common internal modification on eukaryotic mRNA, which has attracted increasing attention in recent years. Accumulating studies have shown that aberrant m6A modifications have profound effects on the characteristics of tumors, which undoubtedly led to a significant breakthrough in cancer treatment. Although m6A function as an oncogene or tumor suppressor is not fully revealed, determining its precise function in the development and evolution of malignant tumors is crucial in improving clinical decisions involving targeted therapies. In this review, we briefly introduce the composition of the m6A methylation machinery and mainly summarize the biological mechanism of m6A in cancer cell death, angiogenesis, epithelial-mesenchymal transition (EMT), and therapeutic resistance. Subsequently, we present the exogenous regulatory factors of m6A and highlight the role of m6A on immune cells and cancer immunotherapy. The potential therapeutic strategies of m6A in human cancer are also discussed, considering research gaps and future applications.
Collapse
Affiliation(s)
- Hui Li
- Harbin Medical University, 157 Baojian Road, Harbin 150086, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150081, China.,Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150086, China
| | - Hao Wu
- Harbin Medical University, 157 Baojian Road, Harbin 150086, China.,Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150081, China.,Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150086, China
| | - Qin Wang
- Harbin Medical University, 157 Baojian Road, Harbin 150086, China.,Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150081, China.,Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150086, China
| | - Shipeng Ning
- Harbin Medical University, 157 Baojian Road, Harbin 150086, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150081, China.,Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150086, China
| | - Shouping Xu
- Harbin Medical University, 157 Baojian Road, Harbin 150086, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150081, China.,Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150086, China
| | - Da Pang
- Harbin Medical University, 157 Baojian Road, Harbin 150086, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150081, China.,Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150081, China.,Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150086, China
| |
Collapse
|
41
|
Du W, Ren L, Hamblin MH, Fan Y. Endothelial Cell Glucose Metabolism and Angiogenesis. Biomedicines 2021; 9:biomedicines9020147. [PMID: 33546224 PMCID: PMC7913320 DOI: 10.3390/biomedicines9020147] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/31/2021] [Accepted: 01/31/2021] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis, a process of new blood vessel formation from the pre-existing vascular bed, is a critical event in various physiological and pathological settings. Over the last few years, the role of endothelial cell (EC) metabolism in angiogenesis has received considerable attention. Accumulating studies suggest that ECs rely on aerobic glycolysis, rather than the oxidative phosphorylation pathway, to produce ATP during angiogenesis. To date, numerous critical regulators of glucose metabolism, fatty acid oxidation, and glutamine metabolism have been identified to modulate the EC angiogenic switch and pathological angiogenesis. The unique glycolytic feature of ECs is critical for cell proliferation, migration, and responses to environmental changes. In this review, we provide an overview of recent EC glucose metabolism studies, particularly glycolysis, in quiescent and angiogenic ECs. We also summarize and discuss potential therapeutic strategies that take advantage of EC metabolism. The elucidation of metabolic regulation and the precise underlying mechanisms could facilitate drug development targeting EC metabolism to treat angiogenesis-related diseases.
Collapse
Affiliation(s)
- Wa Du
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (W.D.); (L.R.)
| | - Lu Ren
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (W.D.); (L.R.)
| | - Milton H. Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Yanbo Fan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (W.D.); (L.R.)
- Department of Internal Medicine, Division of Cardiovascular Health and Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence:
| |
Collapse
|
42
|
Selenium nanoparticles reduce glucose metabolism and promote apoptosis of glioma cells through reactive oxygen species-dependent manner. Neuroreport 2021; 31:226-234. [PMID: 31876687 DOI: 10.1097/wnr.0000000000001386] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gliomas are the most common, malignant, and lethal tumors in adults. Furthermore, gliomas are highly resistant to current chemotherapeutic drugs. Thus, new effective anticancer drugs for glioma are urgently needed. Selenium nanoparticles have been reported to have potent anti-tumor activity, although the specific mechanism is not fully understood. This study aimed to test the anti-tumor effect of selenium nanoparticles and its mechanism. We used selenium nanoparticles to treat commercial glioma cell lines, and patient-derived glioma cells, and then used the MTT assay to determine selenium nanoparticles effect against these. Apoptotic cell death was determined by annexin V-Fluos staining kit. Glucose uptake, lactate, and adenosine triphosphate production, together with hexokinase 2 and pyruvate kinase activities were measured to determine the glucose metabolism level. Reactive oxygen species production was tested using 2',7'-dichlorodihydrofluorescein diacetate. Our results showed that selenium nanoparticles had a potent cytotoxic effect in glioma cells, regardless of whether they were drug-resistant or not, whereas it showed less toxic effect in normal healthy cells. Further tests showed that selenium nanoparticles treatment leads to apoptotic cell death enhancement and glucose metabolism reduction, and this process was in a reactive oxygen species pathway-dependent manner. These results may provide a novel direction for glioma therapy in the future.
Collapse
|
43
|
The Intratumoral Heterogeneity of Cancer Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1311:149-160. [PMID: 34014541 DOI: 10.1007/978-3-030-65768-0_11] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer is one of the deadliest diseases in the world, causing over half a million deaths a year in the USA alone. Despite recent advances made in the field of cancer biology and the therapies that have been developed [1, 2], it is clear that more advances are necessary for us to classify cancer as curable. The logical question that arises is simple: Why, despite all the technologies and medical innovations of our time, has a complete cure eluded us? This chapter sheds light on one of cancer's most impactful attributes: its heterogeneity and, more specifically, the intratumoral heterogeneity of cancer metabolism. Simply put, what makes cancer one of the deadliest diseases is its ability to change and adapt. Cancer cells' rapid evolution, coupled with their irrepressible ability to divide, gives most of them the advantage over our immune systems. In this chapter, we delve into the complexities of this adaptability and the vital role that metabolism plays in the rise and progression of this heterogeneity.
Collapse
|
44
|
Polychemotherapy with Curcumin and Doxorubicin via Biological Nanoplatforms: Enhancing Antitumor Activity. Pharmaceutics 2020; 12:pharmaceutics12111084. [PMID: 33187385 PMCID: PMC7697177 DOI: 10.3390/pharmaceutics12111084] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/31/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (DOX) is a well-known chemotherapeutic agent extensively applied in the field of cancer therapy. However, similar to other chemotherapeutic agents such as cisplatin, paclitaxel, docetaxel, etoposide and oxaliplatin, cancer cells are able to obtain chemoresistance that limits DOX efficacy. In respect to dose-dependent side effect of DOX, enhancing its dosage is not recommended for effective cancer chemotherapy. Therefore, different strategies have been considered for reversing DOX resistance and diminishing its side effects. Phytochemical are potential candidates in this case due to their great pharmacological activities. Curcumin is a potential antitumor phytochemical isolated from Curcuma longa with capacity of suppressing cancer metastasis and proliferation and affecting molecular pathways. Experiments have demonstrated the potential of curcumin for inhibiting chemoresistance by downregulating oncogene pathways such as MMP-2, TGF-β, EMT, PI3K/Akt, NF-κB and AP-1. Furthermore, coadministration of curcumin and DOX potentiates apoptosis induction in cancer cells. In light of this, nanoplatforms have been employed for codelivery of curcumin and DOX. This results in promoting the bioavailability and internalization of the aforementioned active compounds in cancer cells and, consequently, enhancing their antitumor activity. Noteworthy, curcumin has been applied for reducing adverse effects of DOX on normal cells and tissues via reducing inflammation, oxidative stress and apoptosis. The current review highlights the anticancer mechanism, side effects and codelivery of curcumin and DOX via nanovehicles.
Collapse
|
45
|
Li J, Eu JQ, Kong LR, Wang L, Lim YC, Goh BC, Wong ALA. Targeting Metabolism in Cancer Cells and the Tumour Microenvironment for Cancer Therapy. Molecules 2020; 25:molecules25204831. [PMID: 33092283 PMCID: PMC7588013 DOI: 10.3390/molecules25204831] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
Targeting altered tumour metabolism is an emerging therapeutic strategy for cancer treatment. The metabolic reprogramming that accompanies the development of malignancy creates targetable differences between cancer cells and normal cells, which may be exploited for therapy. There is also emerging evidence regarding the role of stromal components, creating an intricate metabolic network consisting of cancer cells, cancer-associated fibroblasts, endothelial cells, immune cells, and cancer stem cells. This metabolic rewiring and crosstalk with the tumour microenvironment play a key role in cell proliferation, metastasis, and the development of treatment resistance. In this review, we will discuss therapeutic opportunities, which arise from dysregulated metabolism and metabolic crosstalk, highlighting strategies that may aid in the precision targeting of altered tumour metabolism with a focus on combinatorial therapeutic strategies.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK;
| | - Jie Qing Eu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
| | - Li Ren Kong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Yaw Chyn Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Pathology, National University Health System, Singapore 119074, Singapore
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Haematology-Oncology, National University Health System, Singapore 119228, Singapore
| | - Andrea L. A. Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Haematology-Oncology, National University Health System, Singapore 119228, Singapore
- Correspondence: ; Tel.: +65-6779-5555
| |
Collapse
|
46
|
Yetkin-Arik B, Kastelein AW, Klaassen I, Jansen CHJR, Latul YP, Vittori M, Biri A, Kahraman K, Griffioen AW, Amant F, Lok CAR, Schlingemann RO, van Noorden CJF. Angiogenesis in gynecological cancers and the options for anti-angiogenesis therapy. Biochim Biophys Acta Rev Cancer 2020; 1875:188446. [PMID: 33058997 DOI: 10.1016/j.bbcan.2020.188446] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
Abstract
Angiogenesis is required in cancer, including gynecological cancers, for the growth of primary tumors and secondary metastases. Development of anti-angiogenesis therapy in gynecological cancers and improvement of its efficacy have been a major focus of fundamental and clinical research. However, survival benefits of current anti-angiogenic agents, such as bevacizumab, in patients with gynecological cancer, are modest. Therefore, a better understanding of angiogenesis and the tumor microenvironment in gynecological cancers is urgently needed to develop more effective anti-angiogenic therapies, either or not in combination with other therapeutic approaches. We describe the molecular aspects of (tumor) blood vessel formation and the tumor microenvironment and provide an extensive clinical overview of current anti-angiogenic therapies for gynecological cancers. We discuss the different phenotypes of angiogenic endothelial cells as potential therapeutic targets, strategies aimed at intervention in their metabolism, and approaches targeting their (inflammatory) tumor microenvironment.
Collapse
Affiliation(s)
- Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Arnoud W Kastelein
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Charlotte H J R Jansen
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Yani P Latul
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Miloš Vittori
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Aydan Biri
- Department of Obstetrics and Gynecology, Koru Ankara Hospital, Ankara, Turkey
| | - Korhan Kahraman
- Department of Obstetrics and Gynecology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Frederic Amant
- Department of Oncology, KU Leuven, Leuven, Belgium; Center for Gynaecological Oncology, Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Center for Gynaecological Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Center for Gynaecological Oncology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Christianne A R Lok
- Center for Gynaecological Oncology, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Cornelis J F van Noorden
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| |
Collapse
|
47
|
Zhang YH, Yan XZ, Xu SF, Pang ZQ, Li LB, Yang Y, Fan YG, Wang Z, Yu X, Guo C, Ao Q. α-Lipoic Acid Maintains Brain Glucose Metabolism via BDNF/TrkB/HIF-1α Signaling Pathway in P301S Mice. Front Aging Neurosci 2020; 12:262. [PMID: 32973490 PMCID: PMC7471806 DOI: 10.3389/fnagi.2020.00262] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/31/2020] [Indexed: 12/30/2022] Open
Abstract
The microtubule-associated protein tau is closely correlated with hypometabolism in Alzheimer’s disease (AD). α-lipoic acid (LA), which is a naturally occurring cofactor in mitochondrial, has been shown to have properties that can inhibit the tau pathology and neuronal damage in our previous research. However, if LA affects glucose metabolism when it reverses tau pathology remains unclear, especially concerning the potential mechanism. Therefore, we make a further study using the P301S mouse model (a tauopathy and AD mouse model which overexpressing fibrillary tau) to gain a clear idea of the aforementioned problems. Here, we found chronic LA administration significantly increased glucose availability by elevating glucose transporter 3 (GLUT3), GLUT4, vascular endothelial growth factor (VEGF) protein and mRNA level, and heme oxygenase-1 (HO-1) protein level in P301S mouse brains. Meanwhile, we found that LA also promoted glycolysis by directly upregulating hexokinase (HK) activity, indirectly by increasing proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and DNA repair enzymes (OGG1/2 and MTH1). Further, we found the underlying mechanism of restored glucose metabolism might involve in the activation of brain-derived neurotrophic factor (BDNF)/tyrosine Kinase receptor B (TrkB)/hypoxia-inducible factor-1α (HIF-1α) signaling pathway by LA treatment.
Collapse
Affiliation(s)
- Yan-Hui Zhang
- School of Fundamental Sciences, China Medical University, Shenyang, China
| | - Xin-Zhu Yan
- School of Fundamental Sciences, China Medical University, Shenyang, China
| | - Shuang-Feng Xu
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Zhong-Qiu Pang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Lin-Bo Li
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yang Yang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yong-Gang Fan
- Institute of Health Science, China Medical University, Shenyang, China
| | - Zhuo Wang
- Institute of Health Science, China Medical University, Shenyang, China
| | - Xin Yu
- Institute of Health Science, China Medical University, Shenyang, China
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Qiang Ao
- School of Fundamental Sciences, China Medical University, Shenyang, China
| |
Collapse
|
48
|
Wang X, Guo K, Huang B, Lin Z, Cai Z. Role of Glucose Transporters in Drug Membrane Transport. Curr Drug Metab 2020; 21:947-958. [PMID: 32778021 DOI: 10.2174/1389200221666200810125924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 06/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glucose is the main energy component of cellular activities. However, as a polar molecule, glucose cannot freely pass through the phospholipid bilayer structure of the cell membrane. Thus, glucose must rely on specific transporters in the membrane. Drugs with a similar chemical structure to glucose may also be transported through this pathway. METHODS This review describes the structure, distribution, action mechanism and influencing factors of glucose transporters and introduces the natural drugs mediated by these transporters and drug design strategies on the basis of this pathway. RESULTS The glucose transporters involved in glucose transport are of two major types, namely, Na+-dependent and Na+-independent transporters. Glucose transporters can help some glycoside drugs cross the biological membrane. The transmembrane potential is influenced by the chemical structure of drugs. Glucose can be used to modify drugs and improve their ability to cross biological barriers. CONCLUSION The membrane transport mechanism of some glycoside drugs may be related to glucose transporters. Glucose modification may improve the oral bioavailability of drugs or achieve targeted drug delivery.
Collapse
Affiliation(s)
- Xin Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Kunkun Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Baolin Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Zimin Lin
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Zheng Cai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
49
|
Han W, Shi J, Cao J, Dong B, Guan W. Emerging Roles and Therapeutic Interventions of Aerobic Glycolysis in Glioma. Onco Targets Ther 2020; 13:6937-6955. [PMID: 32764985 PMCID: PMC7371605 DOI: 10.2147/ott.s260376] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022] Open
Abstract
Glioma is the most common type of intracranial malignant tumor, with a great recurrence rate due to its infiltrative growth, treatment resistance, intra- and intertumoral genetic heterogeneity. Recently, accumulating studies have illustrated that activated aerobic glycolysis participated in various cellular and clinical activities of glioma, thus influencing the efficacy of radiotherapy and chemotherapy. However, the glycolytic process is too complicated and ambiguous to serve as a novel therapy for glioma. In this review, we generalized the implication of key enzymes, glucose transporters (GLUTs), signalings and transcription factors in the glycolytic process of glioma. In addition, we summarized therapeutic interventions via the above aspects and discussed promising clinical applications for glioma.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Jia Shi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Jiachao Cao
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Bo Dong
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Wei Guan
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| |
Collapse
|
50
|
Veys K, Fan Z, Ghobrial M, Bouché A, García-Caballero M, Vriens K, Conchinha NV, Seuwen A, Schlegel F, Gorski T, Crabbé M, Gilardoni P, Ardicoglu R, Schaffenrath J, Casteels C, De Smet G, Smolders I, Van Laere K, Abel ED, Fendt SM, Schroeter A, Kalucka J, Cantelmo AR, Wälchli T, Keller A, Carmeliet P, De Bock K. Role of the GLUT1 Glucose Transporter in Postnatal CNS Angiogenesis and Blood-Brain Barrier Integrity. Circ Res 2020; 127:466-482. [PMID: 32404031 PMCID: PMC7386868 DOI: 10.1161/circresaha.119.316463] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: Endothelial cells (ECs) are highly glycolytic and generate the majority of their energy via the breakdown of glucose to lactate. At the same time, a main role of ECs is to allow the transport of glucose to the surrounding tissues. GLUT1 (glucose transporter isoform 1/Slc2a1) is highly expressed in ECs of the central nervous system (CNS) and is often implicated in blood-brain barrier (BBB) dysfunction, but whether and how GLUT1 controls EC metabolism and function is poorly understood. Objective: We evaluated the role of GLUT1 in endothelial metabolism and function during postnatal CNS development as well as at the adult BBB. Methods and Results: Inhibition of GLUT1 decreases EC glucose uptake and glycolysis, leading to energy depletion and the activation of the cellular energy sensor AMPK (AMP-activated protein kinase), and decreases EC proliferation without affecting migration. Deletion of GLUT1 from the developing postnatal retinal endothelium reduces retinal EC proliferation and lowers vascular outgrowth, without affecting the number of tip cells. In contrast, in the brain, we observed a lower number of tip cells in addition to reduced brain EC proliferation, indicating that within the CNS, organotypic differences in EC metabolism exist. Interestingly, when ECs become quiescent, endothelial glycolysis is repressed, and GLUT1 expression increases in a Notch-dependent fashion. GLUT1 deletion from quiescent adult ECs leads to severe seizures, accompanied by neuronal loss and CNS inflammation. Strikingly, this does not coincide with BBB leakiness, altered expression of genes crucial for BBB barrier functioning nor reduced vascular function. Instead, we found a selective activation of inflammatory and extracellular matrix related gene sets. Conclusions: GLUT1 is the main glucose transporter in ECs and becomes uncoupled from glycolysis during quiescence in a Notch-dependent manner. It is crucial for developmental CNS angiogenesis and adult CNS homeostasis but does not affect BBB barrier function.
Collapse
Affiliation(s)
- Koen Veys
- From the Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), KU Leuven.,Laboratory of Angiogenesis and Vascular Metabolism (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), Center for Cancer Biology, VIB, Leuven
| | - Zheng Fan
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETHZ) Zurich (Z.F., M.G., T.G., P.G., R.A., K.D.B.)
| | - Moheb Ghobrial
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETHZ) Zurich (Z.F., M.G., T.G., P.G., R.A., K.D.B.).,Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, University of Zurich (UZH) and ETHZ and Division of Neurosurgery, USZ, Zurich (M.G., T.W.)
| | - Ann Bouché
- From the Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), KU Leuven.,Laboratory of Angiogenesis and Vascular Metabolism (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), Center for Cancer Biology, VIB, Leuven
| | - Melissa García-Caballero
- From the Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), KU Leuven.,Laboratory of Angiogenesis and Vascular Metabolism (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), Center for Cancer Biology, VIB, Leuven
| | - Kim Vriens
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology (K. Vriens, S.-M.F.), KU Leuven.,Laboratory of Cellular Metabolism and Metabolic Regulation (K. Vriens, S.-M.F.), Center for Cancer Biology, VIB, Leuven
| | - Nadine Vasconcelos Conchinha
- From the Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), KU Leuven.,Laboratory of Angiogenesis and Vascular Metabolism (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), Center for Cancer Biology, VIB, Leuven
| | - Aline Seuwen
- Institute for Biomedical Engineering (A. Seuwen, F.S., A. Schroeter), UZH/ETHZ, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, UZH, Zurich, Switzerland (A. Seuwen, F.S., A. Schroeter)
| | - Felix Schlegel
- Institute for Biomedical Engineering (A. Seuwen, F.S., A. Schroeter), UZH/ETHZ, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, UZH, Zurich, Switzerland (A. Seuwen, F.S., A. Schroeter)
| | - Tatiane Gorski
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETHZ) Zurich (Z.F., M.G., T.G., P.G., R.A., K.D.B.)
| | - Melissa Crabbé
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, University Hospitals Leuven, Belgium (M.C., C.C., K.V.L.).,Molecular Small Animal Imaging Centre, KU Leuven (M.C., C.C., K.V.L.)
| | - Paola Gilardoni
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETHZ) Zurich (Z.F., M.G., T.G., P.G., R.A., K.D.B.)
| | - Raphaela Ardicoglu
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETHZ) Zurich (Z.F., M.G., T.G., P.G., R.A., K.D.B.)
| | - Johanna Schaffenrath
- Neuroscience Center Zurich (J.S., A.K.), UZH/ETHZ, Zurich, Switzerland.,Department of Neurosurgery, Clinical Neurocentre, USZ, Zurich (J.S., A.K.)
| | - Cindy Casteels
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, University Hospitals Leuven, Belgium (M.C., C.C., K.V.L.).,Molecular Small Animal Imaging Centre, KU Leuven (M.C., C.C., K.V.L.)
| | - Gino De Smet
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences, Vrije Universiteit Brussel (G.D.S., I.S.)
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences, Vrije Universiteit Brussel (G.D.S., I.S.)
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, University Hospitals Leuven, Belgium (M.C., C.C., K.V.L.).,Molecular Small Animal Imaging Centre, KU Leuven (M.C., C.C., K.V.L.)
| | - E Dale Abel
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETHZ) Zurich (Z.F., M.G., T.G., P.G., R.A., K.D.B.).,Fraternal Order of Eagles Diabetes Research Center (E.D.A.), University of Iowa.,Division of Endocrinology and Metabolism, Carver College of Medicine (E.D.A.), University of Iowa
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology (K. Vriens, S.-M.F.), KU Leuven.,Laboratory of Cellular Metabolism and Metabolic Regulation (K. Vriens, S.-M.F.), Center for Cancer Biology, VIB, Leuven
| | - Aileen Schroeter
- Institute for Biomedical Engineering (A. Seuwen, F.S., A. Schroeter), UZH/ETHZ, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, UZH, Zurich, Switzerland (A. Seuwen, F.S., A. Schroeter)
| | - Joanna Kalucka
- From the Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), KU Leuven.,Laboratory of Angiogenesis and Vascular Metabolism (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), Center for Cancer Biology, VIB, Leuven.,Aarhus Institute of advanced studies (AIAS) and Department of Biomedicine, Aarhus University (J.K.)
| | - Anna Rita Cantelmo
- From the Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), KU Leuven.,Laboratory of Angiogenesis and Vascular Metabolism (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), Center for Cancer Biology, VIB, Leuven.,Université de Lille, INSERM U1003, Physiologie Cellulaire, France (A.R.C.)
| | - Thomas Wälchli
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, University of Zurich (UZH) and ETHZ and Division of Neurosurgery, USZ, Zurich (M.G., T.W.).,Group of Brain Vasculature and Neurovascular Unit, Department of Clinical Neurosciences, University Hospital Geneva (T.W.).,Department of Fundamental Neurobiology, Krembil Research Institute (T.W.), Toronto Western Hospital, University Health Network, University of Toronto.,Division of Neurosurgery, Department of Surgery (T.W.), Toronto Western Hospital, University Health Network, University of Toronto
| | - Annika Keller
- Neuroscience Center Zurich (J.S., A.K.), UZH/ETHZ, Zurich, Switzerland.,Department of Neurosurgery, Clinical Neurocentre, USZ, Zurich (J.S., A.K.)
| | - Peter Carmeliet
- From the Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), KU Leuven.,Laboratory of Angiogenesis and Vascular Metabolism (K. Veys, A.B., M.G.-C., N.V.C., J.K., A.R.C., P.C.), Center for Cancer Biology, VIB, Leuven
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETHZ) Zurich (Z.F., M.G., T.G., P.G., R.A., K.D.B.)
| |
Collapse
|