1
|
Lidonnici J, Oberkersch RE. Reciprocal Dynamics of Metabolism and mRNA Translation in Tumor Angiogenesis. Int J Mol Sci 2024; 25:11284. [PMID: 39457064 PMCID: PMC11508371 DOI: 10.3390/ijms252011284] [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: 09/26/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Angiogenesis, the process of formation of new blood vessels from pre-existing vasculature, is essential for tumor growth and metastasis. Anti-angiogenic treatment targeting vascular endothelial growth factor (VEGF) signaling is a powerful tool to combat tumor growth; however, anti-tumor angiogenesis therapy has shown limited efficacy, with survival benefits ranging from only a few weeks to months. Compensation by upregulation of complementary growth factors and switches to different modes of vascularization have made these types of therapies less effective. Recent evidence suggests that targeting specific players in endothelial metabolism is a valuable therapeutic strategy against tumor angiogenesis. Although it is clear that metabolism can modulate the translational machinery, the reciprocal relationship between metabolism and mRNA translational control during tumor angiogenesis is not fully understood. In this review, we explore emerging examples of how endothelial cell metabolism affects mRNA translation during the formation of blood vessels. A deeper comprehension of these mechanisms could lead to the development of innovative therapeutic strategies for both physiological and pathological angiogenesis.
Collapse
Affiliation(s)
- Jacopo Lidonnici
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, 35128 Padova, Italy;
| | | |
Collapse
|
2
|
Maurya M, Liu CH, Bora K, Kushwah N, Pavlovich MC, Wang Z, Chen J. Animal Models of Retinopathy of Prematurity: Advances and Metabolic Regulators. Biomedicines 2024; 12:1937. [PMID: 39335451 PMCID: PMC11428941 DOI: 10.3390/biomedicines12091937] [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: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/30/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a primary cause of visual impairment and blindness in premature newborns, characterized by vascular abnormalities in the developing retina, with microvascular alteration, neovascularization, and in the most severe cases retinal detachment. To elucidate the pathophysiology and develop therapeutics for ROP, several pre-clinical experimental models of ROP were developed in different species. Among them, the oxygen-induced retinopathy (OIR) mouse model has gained the most popularity and critically contributed to our current understanding of pathological retinal angiogenesis and the discovery of potential anti-angiogenic therapies. A deeper comprehension of molecular regulators of OIR such as hypoxia-inducible growth factors including vascular endothelial growth factors as primary perpetrators and other new metabolic modulators such as lipids and amino acids influencing pathological retinal angiogenesis is also emerging, indicating possible targets for treatment strategies. This review delves into the historical progressions that gave rise to the modern OIR models with a focus on the mouse model. It also reviews the fundamental principles of OIR, recent advances in its automated assessment, and a selected summary of metabolic investigation enabled by OIR models including amino acid transport and metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
3
|
Wu Y, Huang J, Liu L, Zhang X, Zhang W, Li Q. CircHIPK3/miR-124 affects angiogenesis in early-onset preeclampsia via CPT1A-mediated fatty acid oxidation. J Mol Med (Berl) 2024; 102:1037-1049. [PMID: 38904677 DOI: 10.1007/s00109-024-02461-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/22/2024] [Accepted: 06/07/2024] [Indexed: 06/22/2024]
Abstract
Multiple theories have been proposed to explain the pathogenesis of early-onset preeclampsia (EOPE), and angiogenic dysfunction is an important part of this pathogenesis. Carnitine palmitoyltransferase (CPT1A) is a key rate-limiting enzyme in the metabolic process of fatty acid oxidation (FAO). FAO regulates endothelial cell (EC) proliferation during vascular germination and is also essential for ab initio deoxyribonucleotide synthesis, but its role in EOPE needs to be further elucidated. In the present study, we investigated its functional role in EOPE by targeting the circHIPK3/miR-124-3p/CPT1A axis. In our study, reduced expression of circHIPK3 and CPT1A and increased expression of miR-124-3p in placental tissues from patients with EOPE were associated with EC dysfunction. Here, we confirmed that CPT1A regulates fatty acid oxidative activity, cell proliferation, and tube formation in ECs by regulating FAO. Functionally, knockdown of circHIPK3 suppressed EC angiogenesis by inhibiting CPT1A-mediated fatty acid oxidative activity, which was ameliorated by CPT1A overexpression. In addition, circHIPK3 regulates CPT1A expression by sponging miR-124-3p. Hence, circHIPK3 knockdown reduced fatty acid oxidation in ECs by sponging miR-124-3p in a CPT1A-dependent manner and inhibited EC proliferation and tube formation, which may have led to aberrant angiogenesis in EOPE. Thus, strategies targeting CPT1A-driven FAO may be promising approaches for the treatment of EOPE. KEY MESSAGES: Decreased Carnitine palmitoyltransferase (CPT1A) expression in preeclampsia(PE). CPT1A overexpression promotes FAO activity and tube formation in ECs. CircHIPK3 can affect CPT1A expression and impaire angiogenesis of EOPE. CircHIPK3 regulates CPT1A expression by acting as a ceRNA of miR-124-3p in HUVECs. Confirming the effect of circHIPK3/miR-124-3p/CPT1A axis on EOPE.
Collapse
Affiliation(s)
- Yanying Wu
- Department of Obstetrics, Xiangya Hospital Central South University, Changsha, China
| | - Jingrui Huang
- Department of Obstetrics, Xiangya Hospital Central South University, Changsha, China
- Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, China
| | - Lijuan Liu
- Department of Obstetrics, Xiangya Hospital Central South University, Changsha, China
| | - Xiaowen Zhang
- Department of Obstetrics, Xiangya Hospital Central South University, Changsha, China
| | - Weishe Zhang
- Department of Obstetrics, Xiangya Hospital Central South University, Changsha, China.
- Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, China.
| | - Qi Li
- Institute of Reproductive Medicine Center, Xiangya Hospital Central South University, Changsha, China.
| |
Collapse
|
4
|
da Silva Costa SM, Ito MT, da Cruz PRS, De Souza BB, Rios VM, Bertozzo VDHE, Camargo ACL, Viturino MGM, Lanaro C, de Albuquerque DM, do Canto AM, Saad STO, Ospina-Prieto S, Ozelo MC, Costa FF, de Melo MB. The molecular mechanism responsible for HbSC retinopathy may depend on the action of the angiogenesis-related genes ROBO1 and SLC38A5. Exp Biol Med (Maywood) 2024; 249:10070. [PMID: 39114443 PMCID: PMC11303203 DOI: 10.3389/ebm.2024.10070] [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: 12/06/2023] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
HbSC disease, a less severe form of sickle cell disease, affects the retina more frequently and patients have higher rates of proliferative retinopathy that can progress to vision loss. This study aimed to identify differences in the expression of endothelial cell-derived molecules associated with the pathophysiology of proliferative sickle cell retinopathy (PSCR). RNAseq was used to compare the gene expression profile of circulating endothelial colony-forming cells from patients with SC hemoglobinopathy and proliferative retinopathy (n = 5), versus SC patients without retinopathy (n = 3). Real-time polymerase chain reaction (qRT-PCR) was used to validate the RNAseq results. A total of 134 differentially expressed genes (DEGs) were found. DEGs were mainly associated with vasodilatation, type I interferon signaling, innate immunity and angiogenesis. Among the DEGs identified, we highlight the most up-regulated genes ROBO1 (log2FoldChange = 4.32, FDR = 1.35E-11) and SLC38A5 (log2FoldChange = 3.36 FDR = 1.59E-07). ROBO1, an axon-guided receptor, promotes endothelial cell migration and contributes to the development of retinal angiogenesis and pathological ocular neovascularization. Endothelial SLC38A5, an amino acid (AA) transporter, regulates developmental and pathological retinal angiogenesis by controlling the uptake of AA nutrient, which may serve as metabolic fuel for the proliferation of endothelial cells (ECs) and consequent promotion of angiogenesis. Our data provide an important step towards elucidating the molecular pathophysiology of PSCR that may explain the differences in ocular manifestations between individuals with hemoglobinopathies and afford insights for new alternative strategies to inhibit pathological angiogenesis.
Collapse
Affiliation(s)
| | - Mirta Tomie Ito
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | | | - Bruno Batista De Souza
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | - Vinicius Mandolesi Rios
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | - Victor de Haidar e Bertozzo
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | - Ana Carolina Lima Camargo
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| | | | - Carolina Lanaro
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | | | - Amanda Morato do Canto
- Departamento de Medicina Translacional, Faculdade de Ciências Médicas, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | | | - Stephanie Ospina-Prieto
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | - Margareth Castro Ozelo
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | - Fernando Ferreira Costa
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas—UNICAMP, Campinas, Brazil
| | - Mônica Barbosa de Melo
- Center for Molecular Biology and Genetic Engineering, State University of Campinas—UNICAMP, Campinas, Brazil
| |
Collapse
|
5
|
Nagarkoti S, Kim YM, Das A, Ash D, A Vitriol E, Read TA, Sudhahar V, Hossain MS, Yadav S, McMenamin M, Kelley S, Lucas R, Stepp D, Belin de Chantemele EJ, Caldwell RB, Fulton DJ, Fukai T, Ushio-Fukai M. Endothelial Drp1 Couples VEGF-induced Redox Signaling with Glycolysis Through Cysteine Oxidation to Drive Angiogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.15.599174. [PMID: 38915542 PMCID: PMC11195263 DOI: 10.1101/2024.06.15.599174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Angiogenesis plays a vital role for postnatal development and tissue repair following ischemia. Reactive oxygen species (ROS) generated by NADPH oxidases (NOXes) and mitochondria act as signaling molecules that promote angiogenesis in endothelial cells (ECs) which mainly relies on aerobic glycolysis for ATP production. However, the connections linking redox signaling with glycolysis are not well understood. The GTPase Drp1 is a member of the dynamin superfamily that moves from cytosol to mitochondria through posttranslational modifications to induce mitochondrial fission. The role of Drp1 in ROS-dependent VEGF signaling and angiogenesis in ECs has not been previously described. Here, we identify an unexpected function of endothelial Drp1 as a redox sensor, transmitting VEGF-induced H 2 O 2 signals to enhance glycolysis and angiogenesis. Loss of Drp1 expression in ECs inhibited VEGF-induced angiogenic responses. Mechanistically, VEGF rapidly induced the NOX4-dependent sulfenylation (CysOH) of Drp1 on Cys 644 , promoting disulfide bond formation with the metabolic kinase AMPK and subsequent sulfenylation of AMPK at Cys 299 / 304 via the mitochondrial fission-mitoROS axis. This cysteine oxidation of AMPK, in turn, enhanced glycolysis and angiogenesis. In vivo , mice with EC-specific Drp1 deficiency or CRISPR/Cas9-engineered "redox-dead" (Cys to Ala) Drp1 knock-in mutations exhibited impaired retinal angiogenesis and post-ischemic neovascularization. Our findings uncover a novel role for endothelial Drp1 in linking VEGF-induced mitochondrial redox signaling to glycolysis through a cysteine oxidation-mediated Drp1-AMPK redox relay, driving both developmental and reparative angiogenesis.
Collapse
|
6
|
Liu Y, Wu Z, Li Y, Chen Y, Zhao X, Wu M, Xia Y. Metabolic reprogramming and interventions in angiogenesis. J Adv Res 2024:S2090-1232(24)00178-4. [PMID: 38704087 DOI: 10.1016/j.jare.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
7
|
Sadeghsoltani F, Hassanpour P, Safari MM, Haiaty S, Rahbarghazi R, Rahmati M, Mota A. Angiogenic activity of mitochondria; beyond the sole bioenergetic organelle. J Cell Physiol 2024; 239:e31185. [PMID: 38219050 DOI: 10.1002/jcp.31185] [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: 10/09/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/15/2024]
Abstract
Angiogenesis is a complex process that involves the expansion of the pre-existing vascular plexus to enhance oxygen and nutrient delivery and is stimulated by various factors, including hypoxia. Since the process of angiogenesis requires a lot of energy, mitochondria play an important role in regulating and promoting this phenomenon. Besides their roles as an oxidative metabolism base, mitochondria are potential bioenergetics organelles to maintain cellular homeostasis via sensing alteration in oxygen levels. Under hypoxic conditions, mitochondria can regulate angiogenesis through different factors. It has been indicated that unidirectional and bidirectional exchange of mitochondria or their related byproducts between the cells is orchestrated via different intercellular mechanisms such as tunneling nanotubes, extracellular vesicles, and gap junctions to maintain the cell homeostasis. Even though, the transfer of mitochondria is one possible mechanism by which cells can promote and regulate the process of angiogenesis under reperfusion/ischemia injury. Despite the existence of a close relationship between mitochondrial donation and angiogenic response in different cell types, the precise molecular mechanisms associated with this phenomenon remain unclear. Here, we aimed to highlight the possible role of mitochondria concerning angiogenesis, especially the role of mitochondrial transport and the possible relation of this transfer with autophagy, the housekeeping phenomenon of cells, and angiogenesis.
Collapse
Affiliation(s)
- Fatemeh Sadeghsoltani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Hassanpour
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mir-Meghdad Safari
- Open Heart ICU of Shahid Madani Cardiovascular Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanya Haiaty
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohamad Rahmati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mota
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Xu M, He Y, Li Y, Liu K, Zhang Y, Su T, Yao Y, Jin X, Zhang X, Lu F. Combined Use of Autologous Sustained-Release Scaffold of Adipokines and Acellular Adipose Matrix to Construct Vascularized Adipose Tissue. Plast Reconstr Surg 2024; 153:348e-360e. [PMID: 37171265 DOI: 10.1097/prs.0000000000010649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
BACKGROUND Adipose tissue engineering plays a key role in the reconstruction of soft-tissue defects. The acellular adipose matrix (AAM) is a promising biomaterial for the construction of engineered adipose tissue. However, AAM lacks sufficient adipoinduction potency because of the abundant loss of matrix-bound adipokines during decellularization. METHODS An adipose-derived extracellular matrix collagen scaffold, "adipose collagen fragment" (ACF), was prepared using a novel mechanical method that provides sustained release of adipokines. Here, the authors used label-free proteomics methods to detect the protein components in AAM and ACF. In vivo, ACF was incorporated into AAM or acellular dermal matrix and implanted into nude mice to evaluate adipogenesis. Neoadipocytes, neovessels, and corresponding gene expression were evaluated. The effects of ACF on adipogenic differentiation of human adipose-derived stem cells and tube formation by human umbilical vein endothelial cells were tested in vitro. RESULTS Proteomics analysis showed that ACF contains diverse adipogenic and angiogenic proteins. ACF can release diverse adipokines and induce highly vascularized, mature adipose tissue in AAM, and even in nonadipogenic acellular dermal matrix. Higher expression of adipogenic markers peroxisome proliferator-activated receptor gamma and CCAAT/enhancer-binding protein alpha and greater numbers of tubule structures were observed in ACF-treated groups in vitro. CONCLUSION The combination of ACF and AAM could serve as a novel and promising strategy to construct mature, vascularized adipose tissue for soft-tissue reconstruction. CLINICAL RELEVANCE STATEMENT The combined use of AAM and ACF has been proven to induce a highly vascularized, mature, engineered adipose tissue in the nude mouse model, which may serve as a promising strategy for soft-tissue reconstruction.
Collapse
Affiliation(s)
- Mimi Xu
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Yunfan He
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Yibao Li
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Kaiyang Liu
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Yuchen Zhang
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Ting Su
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Yao Yao
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Xiaoxuan Jin
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Xiangdong Zhang
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Feng Lu
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| |
Collapse
|
9
|
Zodda E, Tura-Ceide O, Mills NL, Tarragó-Celada J, Carini M, Thomson TM, Cascante M. Autonomous metabolic reprogramming and oxidative stress characterize endothelial dysfunction in acute myocardial infarction. eLife 2023; 12:e86260. [PMID: 38014932 PMCID: PMC10871716 DOI: 10.7554/elife.86260] [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: 01/18/2023] [Accepted: 08/01/2023] [Indexed: 11/29/2023] Open
Abstract
Compelling evidence has accumulated on the role of oxidative stress on the endothelial cell (EC) dysfunction in acute coronary syndrome. Unveiling the underlying metabolic determinants has been hampered by the scarcity of appropriate cell models to address cell-autonomous mechanisms of EC dysfunction. We have generated endothelial cells derived from thrombectomy specimens from patients affected with acute myocardial infarction (AMI) and conducted phenotypical and metabolic characterizations. AMI-derived endothelial cells (AMIECs) display impaired growth, migration, and tubulogenesis. Metabolically, AMIECs displayed augmented ROS and glutathione intracellular content, with a diminished glucose consumption coupled to high lactate production. In AMIECs, while PFKFB3 protein levels of were downregulated, PFKFB4 levels were upregulated, suggesting a shunting of glycolysis towards the pentose phosphate pathway, supported by upregulation of G6PD. Furthermore, the glutaminolytic enzyme GLS was upregulated in AMIECs, providing an explanation for the increase in glutathione content. Finally, AMIECs displayed a significantly higher mitochondrial membrane potential than control ECs, which, together with high ROS levels, suggests a coupled mitochondrial activity. We suggest that high mitochondrial proton coupling underlies the high production of ROS, balanced by PPP- and glutaminolysis-driven synthesis of glutathione, as a primary, cell-autonomous abnormality driving EC dysfunction in AMI.
Collapse
Affiliation(s)
- Erika Zodda
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of BarcelonaBarcelonaSpain
- Institute for Molecular Biology of Barcelona, National Research Council (IBMB-CSIC)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-EDH)MadridSpain
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS); University of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)MadridSpain
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and Girona Biomedical Research Institute (IDIBGI)GironaSpain
| | - Nicholas L Mills
- University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburghUnited Kingdom
| | - Josep Tarragó-Celada
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of BarcelonaBarcelonaSpain
| | - Marina Carini
- Department of Pharmaceutical Sciences, Università degli Studi di MilanoMilanItaly
| | - Timothy M Thomson
- Institute for Molecular Biology of Barcelona, National Research Council (IBMB-CSIC)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-EDH)MadridSpain
- Universidad Peruana Cayetano HerediaLimaPeru
| | - Marta Cascante
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-EDH)MadridSpain
- Institute of Biomedicine (IBUB), University of BarcelonaBarcelonaSpain
| |
Collapse
|
10
|
Morales-Guadarrama G, Méndez-Pérez EA, García-Quiroz J, Avila E, Ibarra-Sánchez MJ, Esparza-López J, García-Becerra R, Larrea F, Díaz L. The Inhibition of the FGFR/PI3K/Akt Axis by AZD4547 Disrupts the Proangiogenic Microenvironment and Vasculogenic Mimicry Arising from the Interplay between Endothelial and Triple-Negative Breast Cancer Cells. Int J Mol Sci 2023; 24:13770. [PMID: 37762073 PMCID: PMC10531243 DOI: 10.3390/ijms241813770] [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: 07/15/2023] [Revised: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Vasculogenic mimicry (VM), a process in which aggressive cancer cells form tube-like structures, plays a crucial role in providing nutrients and escape routes. Highly plastic tumor cells, such as those with the triple-negative breast cancer (TNBC) phenotype, can develop VM. However, little is known about the interplay between the cellular components of the tumor microenvironment and TNBC cells' VM capacity. In this study, we analyzed the ability of endothelial and stromal cells to induce VM when interacting with TNBC cells and analyzed the involvement of the FGFR/PI3K/Akt pathway in this process. VM was corroborated using fluorescently labeled TNBC cells. Only endothelial cells triggered VM formation, suggesting a predominant role of paracrine/juxtacrine factors from an endothelial origin in VM development. Via immunocytochemistry, qPCR, and secretome analyses, we determined an increased expression of proangiogenic factors as well as stemness markers in VM-forming cancer cells. Similarly, endothelial cells primed by TNBC cells showed an upregulation of proangiogenic molecules, including FGF, VEGFA, and several inflammatory cytokines. Endothelium-dependent TNBC-VM formation was prevented by AZD4547 or LY294002, strongly suggesting the involvement of the FGFR/PI3K/Akt axis in this process. Given that VM is associated with poor clinical prognosis, targeting FGFR/PI3K/Akt pharmacologically may hold promise for treating and preventing VM in TNBC tumors.
Collapse
Affiliation(s)
- Gabriela Morales-Guadarrama
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - Edgar A. Méndez-Pérez
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - Janice García-Quiroz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - Euclides Avila
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - María J. Ibarra-Sánchez
- Unidad de Bioquímica Dr. Guillermo Soberón Acevedo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico
| | - José Esparza-López
- Unidad de Bioquímica Dr. Guillermo Soberón Acevedo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, Ciudad de México 04510, Mexico
- Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, Ciudad de México 04510, Mexico
| | - Fernando Larrea
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| |
Collapse
|
11
|
Cignarella A, Boscaro C, Albiero M, Bolego C, Barton M. Post-Transcriptional and Epigenetic Regulation of Estrogen Signaling. J Pharmacol Exp Ther 2023; 386:288-297. [PMID: 37391222 DOI: 10.1124/jpet.123.001613] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/17/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023] Open
Abstract
Post-translational and epigenetic regulation are important mechanisms controlling functions of genes and proteins. Although the "classic" estrogen receptors (ERs) have been acknowledged to function in mediating estrogen effects via transcriptional mechanisms, estrogenic agents modulate the turnover of several proteins via post-transcriptional and post-translational pathways including epigenetics. For instance, the metabolic and angiogenic action of G-protein coupled estrogen receptor (GPER) in vascular endothelial cells has been recently elucidated. By interacting with GPER, 17β-estradiol and the GPER agonist G1 enhance endothelial stability of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and capillary tube formation by increasing ubiquitin-specific peptidase 19 levels, thereby reducing PFKFB3 ubiquitination and proteasomal degradation. In addition to ligands, the functional expression and trafficking of ERs can be modulated by post-translational modification, including palmitoylation. MicroRNAs (miRNAs), the most abundant form of endogenous small RNAs in humans, regulate multiple target genes and are at the center of the multi-target regulatory network. This review also discusses the emerging evidence of how miRNAs affect glycolytic metabolism in cancer, as well as their regulation by estrogens. Restoring dysregulated miRNA expression represents a promising strategy to counteract the progression of cancer and other disease conditions. Accordingly, estrogen post-transcriptional regulatory and epigenetic mechanisms represent novel targets for pharmacological and nonpharmacological intervention for the treatment and prevention of hormone-sensitive noncommunicable diseases, including estrogen-sensitive cancers of the reproductive system in women. SIGNIFICANCE STATEMENT: The effects of estrogen are mediated by several mechanisms that are not limited to the transcriptional regulation of target genes. Slowing down the turnover of master regulators of metabolism by estrogens allows cells to rapidly adapt to environmental cues. Identification of estrogen-targeted microRNAs may lead to the development of novel RNA therapeutics that disrupt pathological angiogenesis in estrogen-dependent cancers.
Collapse
Affiliation(s)
- Andrea Cignarella
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Carlotta Boscaro
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Mattia Albiero
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Chiara Bolego
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Matthias Barton
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| |
Collapse
|
12
|
Averchuk AS, Ryazanova MV, Baranich TI, Stavrovskaya AV, Rozanova NA, Novikova SV, Salmina AB. The Neurotoxic Effect of β-Amyloid Is Accompanied by Changes in the Mitochondrial Dynamics and Autophagy in Neurons and Brain Endothelial Cells in the Experimental Model of Alzheimer's Disease. Bull Exp Biol Med 2023; 175:315-320. [PMID: 37561373 DOI: 10.1007/s10517-023-05859-2] [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: 10/19/2022] [Indexed: 08/11/2023]
Abstract
A comparative assessment of the expression of the mitochondrial fission marker Drp1 and the autophagy marker LC3 in neurons and endothelial cells in the hippocampus and entorhinal cortex during progression of cognitive deficit was performed in animals with intrahippocampal administration of β-amyloid. In both brain regions, the expression of Drp1 and LC3 in neuronal and endothelial cells was enhanced. The peak of cognitive impairment corresponded to the maximum expression of Drp1 and LC3 in hippocampal neurons and was preceded by an increase in the number of Drp1+ and LC3+ endothelial cells in this brain region. These data attests to a possible role of aberrant mitochondrial dynamics and autophagy of endothelial cells in the impairment of brain plasticity in the Alzheimer's type neurodegeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | - A B Salmina
- Research Center of Neurology, Moscow, Russia
| |
Collapse
|
13
|
Ruan Q, Tan S, Guo L, Ma D, Wen J. Prevascularization techniques for dental pulp regeneration: potential cell sources, intercellular communication and construction strategies. Front Bioeng Biotechnol 2023; 11:1186030. [PMID: 37274160 PMCID: PMC10232868 DOI: 10.3389/fbioe.2023.1186030] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023] Open
Abstract
One of the difficulties of pulp regeneration is the rapid vascularization of transplanted engineered tissue, which is crucial for the initial survival of the graft and subsequent pulp regeneration. At present, prevascularization techniques, as emerging techniques in the field of pulp regeneration, has been proposed to solve this challenge and have broad application prospects. In these techniques, endothelial cells and pericytes are cocultured to induce intercellular communication, and the cell coculture is then introduced into the customized artificial vascular bed or induced to self-assembly to simulate the interaction between cells and extracellular matrix, which would result in construction of a prevascularization system, preformation of a functional capillary network, and rapid reconstruction of a sufficient blood supply in engineered tissue after transplantation. However, prevascularization techniques for pulp regeneration remain in their infancy, and there remain unresolved problems regarding cell sources, intercellular communication and the construction of prevascularization systems. This review focuses on the recent advances in the application of prevascularization techniques for pulp regeneration, considers dental stem cells as a potential cell source of endothelial cells and pericytes, discusses strategies for their directional differentiation, sketches the mechanism of intercellular communication and the potential application of communication mediators, and summarizes construction strategies for prevascularized systems. We also provide novel ideas for the extensive application and follow-up development of prevascularization techniques for dental pulp regeneration.
Collapse
Affiliation(s)
| | | | | | - Dandan Ma
- *Correspondence: Dandan Ma, ; Jun Wen,
| | - Jun Wen
- *Correspondence: Dandan Ma, ; Jun Wen,
| |
Collapse
|
14
|
Niu N, Ye J, Hu Z, Zhang J, Wang Y. Regulative Roles of Metabolic Plasticity Caused by Mitochondrial Oxidative Phosphorylation and Glycolysis on the Initiation and Progression of Tumorigenesis. Int J Mol Sci 2023; 24:ijms24087076. [PMID: 37108242 PMCID: PMC10139088 DOI: 10.3390/ijms24087076] [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: 02/03/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
One important feature of tumour development is the regulatory role of metabolic plasticity in maintaining the balance of mitochondrial oxidative phosphorylation and glycolysis in cancer cells. In recent years, the transition and/or function of metabolic phenotypes between mitochondrial oxidative phosphorylation and glycolysis in tumour cells have been extensively studied. In this review, we aimed to elucidate the characteristics of metabolic plasticity (emphasizing their effects, such as immune escape, angiogenesis migration, invasiveness, heterogeneity, adhesion, and phenotypic properties of cancers, among others) on tumour progression, including the initiation and progression phases. Thus, this article provides an overall understanding of the influence of abnormal metabolic remodeling on malignant proliferation and pathophysiological changes in carcinoma.
Collapse
Affiliation(s)
- Nan Niu
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
- College of Physics and Optoelectronic Engineering, Canghai Campus of Shenzhen University, Shenzhen 518060, China
| | - Jinfeng Ye
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Zhangli Hu
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Junbin Zhang
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Yun Wang
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
15
|
Chen Y, Tan L, Gao J, Lin C, Wu F, Li Y, Zhang J. Targeting glutaminase 1 (GLS1) by small molecules for anticancer therapeutics. Eur J Med Chem 2023; 252:115306. [PMID: 36996714 DOI: 10.1016/j.ejmech.2023.115306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Glutaminase-1 (GLS1) is a critical enzyme involved in several cellular processes, and its overexpression has been linked to the development and progression of cancer. Based on existing research, GLS1 plays a crucial role in the metabolic activities of cancer cells, promoting rapid proliferation, cell survival, and immune evasion. Therefore, targeting GLS1 has been proposed as a promising cancer therapy strategy, with several GLS1 inhibitors currently under development. To date, several GLS1 inhibitors have been identified, which can be broadly classified into two types: active site and allosteric inhibitors. Despite their pre-clinical effectiveness, only a few number of these inhibitors have advanced to initial clinical trials. Hence, the present medical research emphasizes the need for developing small molecule inhibitors of GLS1 possessing significantly high potency and selectivity. In this manuscript, we aim to summarize the regulatory role of GLS1 in physiological and pathophysiological processes. We also provide a comprehensive overview of the development of GLS1 inhibitors, focusing on multiple aspects such as target selectivity, in vitro and in vivo potency and structure-activity relationships.
Collapse
Affiliation(s)
- Yangyang Chen
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lun Tan
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jing Gao
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Congcong Lin
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Fengbo Wu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yang Li
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
16
|
Furuta K, Tang X, Islam S, Tapia A, Chen ZB, Ibrahim SH. Endotheliopathy in the metabolic syndrome: Mechanisms and clinical implications. Pharmacol Ther 2023; 244:108372. [PMID: 36894027 PMCID: PMC10084912 DOI: 10.1016/j.pharmthera.2023.108372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023]
Abstract
The increasing prevalence of the metabolic syndrome (MetS) is a threat to global public health due to its lethal complications. Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the MetS characterized by hepatic steatosis, which is potentially progressive to the inflammatory and fibrotic nonalcoholic steatohepatitis (NASH). The adipose tissue (AT) is also a major metabolic organ responsible for the regulation of whole-body energy homeostasis, and thereby highly involved in the pathogenesis of the MetS. Recent studies suggest that endothelial cells (ECs) in the liver and AT are not just inert conduits but also crucial mediators in various biological processes via the interaction with other cell types in the microenvironment both under physiological and pathological conditions. Herein, we highlight the current knowledge of the role of the specialized liver sinusoidal endothelial cells (LSECs) in NAFLD pathophysiology. Next, we discuss the processes through which AT EC dysfunction leads to MetS progression, with a focus on inflammation and angiogenesis in the AT as well as on endothelial-to-mesenchymal transition of AT-ECs. In addition, we touch upon the function of ECs residing in other metabolic organs including the pancreatic islet and the gut, the dysregulation of which may also contribute to the MetS. Finally, we highlight potential EC-based therapeutic targets for human MetS, and NASH based on recent achievements in basic and clinical research and discuss how to approach unsolved problems in the field.
Collapse
Affiliation(s)
- Kunimaro Furuta
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA; Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Xiaofang Tang
- Department of Diabetes Complications & Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Shahidul Islam
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Alonso Tapia
- Department of Diabetes Complications & Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Zhen Bouman Chen
- Department of Diabetes Complications & Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Samar H Ibrahim
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA; Division of Pediatric Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
17
|
Brindisi M, Frattaruolo L, Fiorillo M, Dolce V, Sotgia F, Lisanti MP, Cappello AR. New insights into cholesterol-mediated ERRα activation in breast cancer progression and pro-tumoral microenvironment orchestration. FEBS J 2023; 290:1481-1501. [PMID: 36237175 DOI: 10.1111/febs.16651] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 09/08/2022] [Accepted: 10/12/2022] [Indexed: 01/31/2023]
Abstract
Breast cancer remains the greatest cause of cancer-related death in women worldwide. Its aggressiveness and progression derive from intricate processes that occur simultaneously both within the tumour itself and in the neighbouring cells that make up its microenvironment. The aim of the present work was firstly to study how elevated cholesterol levels increase tumour aggressiveness. Herein, we demonstrate that cholesterol, by activating ERRα pathway, promotes epithelium-mesenchymal transition (EMT) in breast cancer cells (MCF-7 and MDA-MB-231) as well as the release of pro-inflammatory factors able to orchestrate the tumour microenvironment. A further objective of this work was to study the close symbiosis between tumour cells and the microenvironment. Our results allow us to highlight, for the first time, that breast cancer cells exposed to high cholesterol levels promote (a) greater macrophages infiltration with induction of an M2 phenotype, (b) angiogenesis and endothelial branching, as well as (c) a cancer-associated fibroblasts (CAFs) phenotype. The effects observed could be due to direct activation of the ERRα pathway by high cholesterol levels, since the simultaneous inhibition of this pathway subverts such effects. Overall, these findings enable us to identify the cholesterol-ERRα synergy as an interesting target for breast cancer treatment.
Collapse
Affiliation(s)
- Matteo Brindisi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
- Cell Adhesion Unit, Vita-Salute San Raffaele University, Milan, Italy
| | - Luca Frattaruolo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
- Translational Medicine, School of Science, Engineering and the Environment (SEE), University of Salford, Greater Manchester, UK
| | - Vincenza Dolce
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Federica Sotgia
- Translational Medicine, School of Science, Engineering and the Environment (SEE), University of Salford, Greater Manchester, UK
| | - Michael P Lisanti
- Translational Medicine, School of Science, Engineering and the Environment (SEE), University of Salford, Greater Manchester, UK
| | - Anna Rita Cappello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
18
|
Jovankić JV, Nikodijević DD, Milutinović MG, Nikezić AG, Kojić VV, Cvetković AM, Cvetković DM. Potential of Orlistat to induce apoptotic and antiangiogenic effects as well as inhibition of fatty acid synthesis in breast cancer cells. Eur J Pharmacol 2023; 939:175456. [PMID: 36528070 DOI: 10.1016/j.ejphar.2022.175456] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/04/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Breast cancer as most often women's cancer is the second cause of mortality worldwide. Research interest increased in testing non-standard drugs to suppress breast cancer progression and become significant supplements in anticancer therapy. The anti-obesity drug Orlistat showed significant ability for modulation of cancer cell metabolism via antiproliferative, proapoptotic, antiangiogenic, antimetastatic, and hypolipidemic effects. The anticancer potential of Orlistat was evaluated by cytotoxicity (MTT assay), type of cell death (AO/EB double staining), determination of redox status parameters (superoxide, hydrogen peroxide, lipid peroxidation, reduced glutathione), and total lipid levels with colorimetric methods, as well on angiogenesis-related (VEGF, MMP-9, CXCR4/CXCL12) and fatty acid synthesis-related (ACLY, ACC, FASN) parameters on gene and protein levels (immunocytochemistry and qPCR). Based on obtained results Orlistat induces significant cytotoxic, proapoptotic, and anti-angiogenic effects in MDA-MB-231, MDA-MB-468 and MCF-7 breast cancer cells, without significant cytotoxic effects on normal MRC-5 cells. It decreased total lipid levels and changed redox status parameters and cancer cell metabolism via suppression of genes and proteins involved and fatty acid synthesis. Based on showed, Orlistat may be an important supplement in antiangiogenic therapy against breast cancer with no side effects on normal cells, making it a good candidate for future clinical trials.
Collapse
Affiliation(s)
- Jovana V Jovankić
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Danijela D Nikodijević
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Milena G Milutinović
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia.
| | - Aleksandra G Nikezić
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Vesna V Kojić
- University of Novi Sad, Faculty of Medicine, Oncology Institute of Vojvodina, Put Dr Goldmana 4, Sremska Kamenica, 21204, Serbia
| | - Aleksandar M Cvetković
- University of Kragujevac, Faculty of Medical Sciences, Department of Surgery, Svetozara Markovića 69, 34000, Kragujevac, Serbia
| | - Danijela M Cvetković
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Natural Sciences, Jovana Cvijića bb, 34000, Kragujevac, Serbia
| |
Collapse
|
19
|
Fossel M, Bean J, Khera N, Kolonin MG. A Unified Model of Age-Related Cardiovascular Disease. BIOLOGY 2022; 11:1768. [PMID: 36552277 PMCID: PMC9775230 DOI: 10.3390/biology11121768] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/18/2022] [Accepted: 11/27/2022] [Indexed: 12/12/2022]
Abstract
Despite progress in biomedical technologies, cardiovascular disease remains the main cause of mortality. This is at least in part because current clinical interventions do not adequately take into account aging as a driver and are hence aimed at suboptimal targets. To achieve progress, consideration needs to be given to the role of cell aging in disease pathogenesis. We propose a model unifying the fundamental processes underlying most age-associated cardiovascular pathologies. According to this model, cell aging, leading to cell senescence, is responsible for tissue changes leading to age-related cardiovascular disease. This process, occurring due to telomerase inactivation and telomere attrition, affects all components of the cardiovascular system, including cardiomyocytes, vascular endothelial cells, smooth muscle cells, cardiac fibroblasts, and immune cells. The unified model offers insights into the relationship between upstream risk factors and downstream clinical outcomes and explains why interventions aimed at either of these components have limited success. Potential therapeutic approaches are considered based on this model. Because telomerase activity can prevent and reverse cell senescence, telomerase gene therapy is discussed as a promising intervention. Telomerase gene therapy and similar systems interventions based on the unified model are expected to be transformational in cardiovascular medicine.
Collapse
Affiliation(s)
| | - Joe Bean
- University of Missouri School of Medicine, Kansas City, MO 65211, USA
| | - Nina Khera
- Buckingham Browne and Nichols School, Wellesley, MA 02138, USA
| | - Mikhail G. Kolonin
- University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
20
|
Wang Z, Yemanyi F, Blomfield AK, Bora K, Huang S, Liu CH, Britton WR, Cho SS, Tomita Y, Fu Z, Ma JX, Li WH, Chen J. Amino acid transporter SLC38A5 regulates developmental and pathological retinal angiogenesis. eLife 2022; 11:e73105. [PMID: 36454214 PMCID: PMC9714971 DOI: 10.7554/elife.73105] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
Amino acid (AA) metabolism in vascular endothelium is important for sprouting angiogenesis. SLC38A5 (solute carrier family 38 member 5), an AA transporter, shuttles neutral AAs across cell membrane, including glutamine, which may serve as metabolic fuel for proliferating endothelial cells (ECs) to promote angiogenesis. Here, we found that Slc38a5 is highly enriched in normal retinal vascular endothelium, and more specifically, in pathological sprouting neovessels. Slc38a5 is suppressed in retinal blood vessels from Lrp5-/- and Ndpy/- mice, both genetic models of defective retinal vascular development with Wnt signaling mutations. Additionally, Slc38a5 transcription is regulated by Wnt/β-catenin signaling. Genetic deficiency of Slc38a5 in mice substantially delays retinal vascular development and suppresses pathological neovascularization in oxygen-induced retinopathy modeling ischemic proliferative retinopathies. Inhibition of SLC38A5 in human retinal vascular ECs impairs EC proliferation and angiogenic function, suppresses glutamine uptake, and dampens vascular endothelial growth factor receptor 2. Together these findings suggest that SLC38A5 is a new metabolic regulator of retinal angiogenesis by controlling AA nutrient uptake and homeostasis in ECs.
Collapse
Affiliation(s)
- Zhongxiao Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Felix Yemanyi
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Alexandra K Blomfield
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Kiran Bora
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Shuo Huang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - William R Britton
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Steve S Cho
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Yohei Tomita
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Jian-xing Ma
- Department of Biochemistry, Wake Forest University School of MedicineWinston-SalemUnited States
| | - Wen-hong Li
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical CenterDallasUnited States
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
21
|
Patil N, Howe O, Cahill P, Byrne HJ. Monitoring and modelling the dynamics of the cellular glycolysis pathway: A review and future perspectives. Mol Metab 2022; 66:101635. [PMID: 36379354 PMCID: PMC9703637 DOI: 10.1016/j.molmet.2022.101635] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The dynamics of the cellular glycolysis pathway underpin cellular function and dysfunction, and therefore ultimately health, disease, diagnostic and therapeutic strategies. Evolving our understanding of this fundamental process and its dynamics remains critical. SCOPE OF REVIEW This paper reviews the medical relevance of glycolytic pathway in depth and explores the current state of the art for monitoring and modelling the dynamics of the process. The future perspectives of label free, vibrational microspectroscopic techniques to overcome the limitations of the current approaches are considered. MAJOR CONCLUSIONS Vibrational microspectroscopic techniques can potentially operate in the niche area of limitations of other omics technologies for non-destructive, real-time, in vivo label-free monitoring of glycolysis dynamics at a cellular and subcellular level.
Collapse
Affiliation(s)
- Nitin Patil
- FOCAS Research Institute, Technological University Dublin, City Campus, Camden Row, Dublin 8, Ireland; School of Physics and Optometric & Clinical Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland.
| | - Orla Howe
- School of Biological and Health Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland
| | - Paul Cahill
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Hugh J Byrne
- FOCAS Research Institute, Technological University Dublin, City Campus, Camden Row, Dublin 8, Ireland
| |
Collapse
|
22
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
23
|
Mohammadi P, Yarani R, Rahimpour A, Ranjbarnejad F, Mendes Lopes de Melo J, Mansouri K. Targeting endothelial cell metabolism in cancerous microenvironment: a new approach for anti-angiogenic therapy. Drug Metab Rev 2022; 54:386-400. [PMID: 36031813 DOI: 10.1080/03602532.2022.2116033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Anti-angiogenic therapy is a practical approach to managing diseases with increased angiogenesis, such as cancer, maculopathies, and retinopathies. Considering the fundamental gaps in the knowledge of the vital pathways involved in angiogenesis and its inhibition and the insufficient efficiency of existing angiogenesis inhibitors, there is an increasing focus on the emergence of new therapeutic strategies aimed at inhibiting pathological angiogenesis. Angiogenesis is forming a new vascular network from existing vessels; endothelial cells (ECs), vascular lining cells, are the main actors of angiogenesis in physiological or pathological conditions. Switching from a quiescent state to a highly migratory and proliferative state during new vessel formation called "angiogenic switch" is driven by a "metabolic switch" in ECs, angiogenic growth factors, and other signals. As the characteristics of ECs change by altering the surrounding environment, they appear to have a different metabolism in a tumor microenvironment (TME). Therefore, pathological angiogenesis can be inhibited by targeting metabolic pathways. In the current review, we aim to discuss the EC metabolic pathways under normal and TME conditions to verify the suitability of targeting them with novel therapies.
Collapse
Affiliation(s)
- Parisa Mohammadi
- Medical Biology Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Azam Rahimpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical, Sciences, Tehran, Iran
| | - Fatemeh Ranjbarnejad
- Medical Biology Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Joana Mendes Lopes de Melo
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Kamran Mansouri
- Medical Biology Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
24
|
Boyang C, Yuexing L, Yiping Y, Haiyang Y, Xufei Z, Liancheng G, Yunzhi C. Construction and analysis of heart failure diagnosis model based on random forest and artificial neural network. Medicine (Baltimore) 2022; 101:e31097. [PMID: 36254001 PMCID: PMC9575800 DOI: 10.1097/md.0000000000031097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Heart failure is a global health problem and the number of sufferers is increasing as the population grows and ages. Existing diagnostic techniques for heart failure have various limitations in the clinical setting and there is a need to develop a new diagnostic model to complement the existing diagnostic methods. In recent years, with the development and improvement of gene sequencing technology, more genes associated with heart failure have been identified. We screened for differentially expressed genes in heart failure using available gene expression data from the Gene Expression Omnibus database and identified 6 important genes by a random forest classifier (ASPN, MXRA5, LUM, GLUL, CNN1, and SERPINA3). And we have successfully constructed a new heart failure diagnostic model using an artificial neural network and validated its diagnostic efficacy in a public dataset. We calculated heart failure-related differentially expressed genes and obtained 24 candidate genes by random forest classification, and selected the top 6 genes as important genes for subsequent analysis. The prediction weights of the genes of interest were determined by the neural network model and the model scores were evaluated in 2 independent sample datasets (GSE16499 and GSE57338 datasets). Since the weights of RNA-seq predictions for constructing neural network models were theoretically more suitable for disease classification of RNA-seq data, the GSE57338 dataset had the best performance in the validation results. The diagnostic model derived from our study can be of clinical value in determining the likelihood of HF occurring through cardiac biopsy. In the meantime, we need to further investigate the accuracy of the diagnostic model based on the results of our study.
Collapse
Affiliation(s)
- Chen Boyang
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Li Yuexing
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yan Yiping
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yu Haiyang
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Zhang Xufei
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Guan Liancheng
- Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Chen Yunzhi
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- * Correspondence: Chen Yunzhi, School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China (e-mail: )
| |
Collapse
|
25
|
Li H, Liu P, Li D, Wang Z, Ding Z, Zhou M, Chen X, Miao M, Ding J, Lin W, Liu Y, Zha X. STAT3/miR-130b-3p/MBNL1 feedback loop regulated by mTORC1 signaling promotes angiogenesis and tumor growth. J Exp Clin Cancer Res 2022; 41:297. [PMID: 36217202 PMCID: PMC9552455 DOI: 10.1186/s13046-022-02513-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Aberrantly activated mammalian target of rapamycin complex 1 (mTORC1) plays a vital role in tumor angiogenesis, but its precise mechanisms are still unclear. METHODS Micro-RNA-130b-3p (miR-130b-3p) expression in mTORC1-activated and control cells was examined by quantitative real-time PCR (qRT-PCR). MiR-130b-3p levels and their correlation with mTORC1 activity were evaluated by analyzing publicly available databases and in-house head and neck squamous cell carcinoma (HNSCC) tissues. The role of miR-130b-3p in mTORC1-mediated angiogenesis and tumor growth was examined using tube formation assay, chicken chorioallantoic membrane assay, cell line - derived xenograft models, and an HNSCC patient-derived xenograft (PDX) model. The regulatory mechanisms among signal transducer and activator of transcription 3 (STAT3), miR-130b-3p, and muscleblind-like protein 1 (MBNL1) were investigated via bioinformatics analyses, qRT-PCR, western blot, RNA immunoprecipitation, immunofluorescence, luciferase reporter assay, and chromatin immunoprecipitation assay. RESULTS Elevated miR-130b-3p enhanced the angiogenic and tumorigenic abilities of mTORC1-activated cells both in vitro and in vivo. STAT3, a downstream effector of mTORC1, transactivated miR-130b-3p by direct binding promoter of the miR-130b gene. MBNL1 was identified as a direct target of miR-130b-3p. MBNL1 depletion rescued the compromised angiogenesis and tumor growth caused by miR-130b-3p inhibition. MiR-130b-3p levels were significantly upregulated and positively correlated with mTORC1 signaling in multiple cancers. MiR-130b-3p inhibition attenuated tumor angiogenesis and growth in an HNSCC PDX model. MBNL1 feedback inhibited STAT3 activation in mTORC1-activated cells. CONCLUSIONS The STAT3/miR-130b-3p/MBNL1 feedback loop plays a vital role in mTORC1-mediated angiogenesis and tumor progression. This pathway could be targeted for therapeutic intervention of mTORC1-related cancers.
Collapse
Affiliation(s)
- Hongwu Li
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- Anhui Public Health Clinical Center, Hefei, 230032, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Ping Liu
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- Anhui Public Health Clinical Center, Hefei, 230032, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Dapeng Li
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zixi Wang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Zhao Ding
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Meng Zhou
- Department of Pharmacy, Genertec Universal Medical Maanshan Shiqiye Hospital, Maanshan, 243000, Anhui Province, China
| | - Xu Chen
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Manli Miao
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Junli Ding
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Wei Lin
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Yehai Liu
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
26
|
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: 15] [Impact Index Per Article: 7.5] [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
|
27
|
Ngwa VM, Edwards DN, Hwang Y, Karno B, Wang X, Yan C, Richmond A, Brantley-Sieders DM, Chen J. Loss of vascular endothelial glutaminase inhibits tumor growth and metastasis, and increases sensitivity to chemotherapy. CANCER RESEARCH COMMUNICATIONS 2022; 2:694-705. [PMID: 36381236 PMCID: PMC9645801 DOI: 10.1158/2767-9764.crc-22-0048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/06/2022] [Accepted: 06/23/2022] [Indexed: 02/02/2023]
Abstract
Glutamine is the most abundant non-essential amino acid in blood stream; yet it's concentration in tumor interstitium is markedly lower than that in the serum, reflecting the huge demand of various cell types in tumor microenvironment for glutamine. While many studies have investigated glutamine metabolism in tumor epithelium and infiltrating immune cells, the role of glutamine metabolism in tumor blood vessels remains unknown. Here, we report that inducible genetic deletion of glutaminase (GLS) specifically in host endothelium, GLSECKO, impairs tumor growth and metastatic dissemination in vivo. Loss of GLS decreased tumor microvascular density, increased perivascular support cell coverage, improved perfusion, and reduced hypoxia in mammary tumors. Importantly, chemotherapeutic drug delivery and therapeutic efficacy were improved in tumor-bearing GLSECKO hosts or in combination with GLS inhibitor, CB839. Mechanistically, loss of GLS in tumor endothelium resulted in decreased leptin levels, and exogenous recombinant leptin rescued tumor growth defects in GLSECKO mice. Together, these data demonstrate that inhibition of endothelial glutamine metabolism normalizes tumor vessels, reducing tumor growth and metastatic spread, improving perfusion, and reducing hypoxia, and enhancing chemotherapeutic delivery. Thus, targeting glutamine metabolism in host vasculature may improve clinical outcome in patients with solid tumors.
Collapse
Affiliation(s)
- Verra M. Ngwa
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Deanna N. Edwards
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee
- Department of Medicine, Division of Rheumatology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yoonha Hwang
- Department of Medicine, Division of Rheumatology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Breelyn Karno
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Xiaoyong Wang
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee
- Department of Medicine, Division of Rheumatology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Chi Yan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ann Richmond
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dana M. Brantley-Sieders
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee
- Department of Medicine, Division of Rheumatology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jin Chen
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee
- Department of Medicine, Division of Rheumatology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
28
|
Moccia F, Negri S, Faris P, Ronchi C, Lodola F. Optical excitation of organic semiconductors as a highly selective strategy to induce vascular regeneration and tissue repair. Vascul Pharmacol 2022; 144:106998. [PMID: 35589009 DOI: 10.1016/j.vph.2022.106998] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/06/2022] [Accepted: 05/12/2022] [Indexed: 10/18/2022]
Abstract
Therapeutic neovascularization represents a promising strategy to rescue the vascular network and restore organ function in cardiovascular disorders (CVDs), including acute myocardial infarction, heart failure, peripheral artery disease, and brain stroke. Endothelial colony forming cells (ECFCs), which are mobilized in circulation upon an ischemic insult, are commonly regarded as the most suitable cellular tool to achieve therapeutic neovascularization. ECFCs can be genetically or pharmacologically manipulated to enhance their vasoreparative potential by boosting specific pro-angiogenic signalling pathways. However, optical stimulation represents the most reliable approach to control cellular activity because of its high selectivity and unprecedented spatio-temporal resolution. Herein, we discuss a novel strategy to drive ECFC angiogenic activity in ischemic tissues by combining geneless optical excitation with photosensitive organic semiconductors. We describe how photoexcitation of the conducting polymer poly(3-hexylthiophene-2,5-diyl), also known as P3HT, stimulates extracellular Ca2+ entry through Transient Receptor Potential Vanilloid 1 (TRPV1) channels upon the production of hydrogen peroxide (H2O2) in the cleft between the nanomaterial and the cell membrane. H2O2-induced TRPV1-dependent Ca2+ entry stimulates ECFC proliferation and tube formation, thereby providing the proof-of-concept that photoexcitation of organic semiconductors may offer a reliable strategy to stimulate ECFCs-dependent neovascularization in CVDs.
Collapse
Affiliation(s)
- Francesco Moccia
- Department of Biology and Biotechnology "Lazzaro Spallanzani", Laboratory of General Physiology, University of Pavia, 27100 Pavia, Italy.
| | - Sharon Negri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", Laboratory of General Physiology, University of Pavia, 27100 Pavia, Italy
| | - Pawan Faris
- Department of Biology and Biotechnology "Lazzaro Spallanzani", Laboratory of General Physiology, University of Pavia, 27100 Pavia, Italy
| | - Carlotta Ronchi
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, 20133 Milan, Italy
| | - Francesco Lodola
- Department of Biotechnology and Bioscience, Laboratory of Cardiac Cellular Physiology, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
29
|
Ando T, Tai-Nagara I, Sugiura Y, Kusumoto D, Okabayashi K, Kido Y, Sato K, Saya H, Navankasattusas S, Li DY, Suematsu M, Kitagawa Y, Seiradake E, Yamagishi S, Kubota Y. Tumor-specific inter-endothelial adhesion mediated by FLRT2 facilitates cancer aggressiveness. J Clin Invest 2022; 132:153626. [PMID: 35104247 PMCID: PMC8920344 DOI: 10.1172/jci153626] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Blood vessel abnormalization alters cancer cell metabolism and promotes cancer dissemination and metastasis. However, the biological features of the abnormalized blood vessels that facilitate cancer progression and whether they can be targeted therapeutically have not been fully investigated. Here, we found that an axon guidance molecule, fibronectin leucine-rich transmembrane protein 2 (FLRT2), is expressed preferentially in abnormalized vessels of advanced colorectal cancers in humans, and that its expression correlates negatively with long-term survival. Endothelial-specific deletion of Flrt2 in mice selectively pruned abnormalized vessels, resulting in a unique metabolic state termed "oxygen-glucose uncoupling", which suppresses tumor metastasis. Moreover, Flrt2 deletion caused an increase in the number of mature vessels, resulting in a significant increase in the anti-tumor effects of immune checkpoint blockers. Mechanistically, we found that FLRT2 forms non-canonical inter-endothelial adhesions that safeguard against oxidative stress through homophilic binding. Together, our results demonstrate the existence of tumor-specific inter-endothelial adhesions that enable abnormalized vessels to facilitate cancer aggressiveness. Targeting this type of adhesion complex could be a safe and effective therapeutic option to suppress cancer progression.
Collapse
Affiliation(s)
- Tomofumi Ando
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ikue Tai-Nagara
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Dai Kusumoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Koji Okabayashi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yasuaki Kido
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kohji Sato
- Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Keio University School of Medicine, Tokyo, Japan
| | - Sutip Navankasattusas
- Department of Medicine, University of Utah, Salt Lake City, United States of America
| | - Dean Y Li
- Department of Medicine, University of Utah, Salt Lake City, United States of America
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Elena Seiradake
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Satoru Yamagishi
- Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
Qiu J, Li Y, Wang B, Sun X, Qian D, Ying Y, Zhou J. The Role and Research Progress of Inhibitor of Differentiation 1 in Atherosclerosis. DNA Cell Biol 2022; 41:71-79. [PMID: 35049366 PMCID: PMC8863915 DOI: 10.1089/dna.2021.0745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022] Open
Abstract
Inhibitor of differentiation 1 has a helix-loop-helix (HLH) structure, belongs to a class of molecules known as the HLH trans-acting factor family, and plays an important role in advancing the cell cycle, promoting cell proliferation and inhibiting cell differentiation. Recent studies have confirmed that inhibitor of differentiation 1 plays an important role in the endothelial-mesenchymal transition of vascular endothelial cells, angiogenesis, reendothelialization after injury, and the formation and rupture of atherosclerotic plaques. An in-depth understanding of the role of inhibitor of differentiation 1 in atherosclerosis will provide new ideas and strategies for the treatment of related diseases.
Collapse
Affiliation(s)
- Jun Qiu
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - Youhong Li
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
| | - BingYu Wang
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - XinYi Sun
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - Dingding Qian
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Yuchen Ying
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Jianqing Zhou
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| |
Collapse
|
31
|
Yang Y, Yang Q, Luo S, Zhang Y, Lian C, He H, Zeng J, Zhang G. Comparative Analysis Reveals Novel Changes in Plasma Metabolites and Metabolomic Networks of Infants With Retinopathy of Prematurity. Invest Ophthalmol Vis Sci 2022; 63:28. [PMID: 35060995 PMCID: PMC8787637 DOI: 10.1167/iovs.63.1.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Advances in mass spectrometry have provided new insights into the role of metabolomics in the etiology of several diseases. Studies on retinopathy of prematurity (ROP), for example, overlooked the role of metabolic alterations in disease development. We employed comprehensive metabolic profiling and gold-standard metabolic analysis to explore major metabolites and metabolic pathways, which were significantly affected in early stages of pathogenesis toward ROP. Methods This was a multicenter, retrospective, matched-pair, case-control study. We collected plasma from 57 ROP cases and 57 strictly matched non-ROP controls. Non-targeted ultra-high-performance liquid chromatography-tandem mass spectroscopy (UPLC-MS/MS) was used to detect the metabolites. Machine learning was employed to reveal the most affected metabolites and pathways in ROP development. Results Compared with non-ROP controls, we found a significant metabolic perturbation in the plasma of ROP cases, which featured an increase in the levels of lipids, nucleotides, and carbohydrate metabolites and lower levels of peptides. Machine leaning enabled us to distinguish a cluster of metabolic pathways (glycometabolism, redox homeostasis, lipid metabolism, and arginine pathway) were strongly correlated with the development of ROP. Moreover, the severity of ROP was associated with the levels of creatinine and ribitol; also, overactivity of aerobic glycolysis and lipid metabolism was noted in the metabolic profile of ROP. Conclusions The results suggest a strong correlation between metabolic profiling and retinal neovascularization in ROP pathogenesis. These findings provide an insight into the identification of novel metabolic biomarkers for the diagnosis and prevention of ROP, but the clinical significance requires further validation.
Collapse
Affiliation(s)
- Yuhang Yang
- Shenzhen Eye Hospital, Shenzhen Key Ophthalmic Laboratory, The Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Qian Yang
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Sisi Luo
- Shenzhen Key Prevention and Control Laboratory of Birth Defects Prevention and Control, Shenzhen Maternal and Child Health Hospital, The Affiliated Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Yinsheng Zhang
- School of Management and E-Business, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Chaohui Lian
- Shenzhen Key Prevention and Control Laboratory of Birth Defects Prevention and Control, Shenzhen Maternal and Child Health Hospital, The Affiliated Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Honghui He
- Shenzhen Eye Hospital, Shenzhen Key Ophthalmic Laboratory, The Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Jian Zeng
- Shenzhen Eye Hospital, Shenzhen Key Ophthalmic Laboratory, The Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Guoming Zhang
- Shenzhen Eye Hospital, Shenzhen Key Ophthalmic Laboratory, The Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| |
Collapse
|
32
|
Yang B, Li G, Liu J, Li X, Zhang S, Sun F, Liu W. Nanotechnology for Age-Related Macular Degeneration. Pharmaceutics 2021; 13:pharmaceutics13122035. [PMID: 34959316 PMCID: PMC8705006 DOI: 10.3390/pharmaceutics13122035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/04/2021] [Accepted: 11/22/2021] [Indexed: 01/12/2023] Open
Abstract
Age-related macular degeneration (AMD) is a degenerative eye disease that is the leading cause of irreversible vision loss in people 50 years and older. Today, the most common treatment for AMD involves repeated intravitreal injections of anti-vascular endothelial growth factor (VEGF) drugs. However, the existing expensive therapies not only cannot cure this disease, they also produce a variety of side effects. For example, the number of injections increases the cumulative risk of endophthalmitis and other complications. Today, a single intravitreal injection of gene therapy products can greatly reduce the burden of treatment and improve visual effects. In addition, the latest innovations in nanotherapy provide the best drug delivery alternative for the treatment of AMD. In this review, we discuss the development of nano-drug delivery systems and gene therapy strategies for AMD in recent years. In addition, we discuss some novel targeting strategies and the potential application of these delivery methods in the treatment of AMD. Finally, we also propose that the combination of CRISPR/Cas9 technology with a new non-viral delivery system may be promising as a therapeutic strategy for the treatment of AMD.
Collapse
Affiliation(s)
- Bo Yang
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun 130012, China;
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Ge Li
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Jiaxin Liu
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Xiangyu Li
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Shixin Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Fengying Sun
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Wenhua Liu
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun 130012, China;
- Correspondence:
| |
Collapse
|
33
|
Bandini E, Rossi T, Scarpi E, Gallerani G, Vannini I, Salvi S, Azzali I, Melloni M, Salucci S, Battistelli M, Serra P, Maltoni R, Cho WC, Fabbri F. Early Detection and Investigation of Extracellular Vesicles Biomarkers in Breast Cancer. Front Mol Biosci 2021; 8:732900. [PMID: 34820420 PMCID: PMC8606536 DOI: 10.3389/fmolb.2021.732900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/07/2021] [Indexed: 02/01/2023] Open
Abstract
Breast cancer (BC) is the most commonly diagnosed malignant tumor in women worldwide, and the leading cause of cancer death in the female population. The percentage of patients experiencing poor prognosis along with the risk of developing metastasis remains high, also affecting the resistance to current main therapies. Cancer progression and metastatic development are no longer due entirely to their intrinsic characteristics, but also regulated by signals derived from cells of the tumor microenvironment. Extracellular vesicles (EVs) packed with DNA, RNA, and proteins, are the most attractive targets for both diagnostic and therapeutic applications, and represent a decisive challenge as liquid biopsy-based markers. Here we performed a study based on a multiplexed phenotyping flow cytometric approach to characterize BC-derived EVs from BC patients and cell lines, through the detection of multiple antigens. Our data reveal the expression of EVs-related biomarkers derived from BC patient plasma and cell line supernatants, suggesting that EVs could be exploited for characterizing and monitoring disease progression.
Collapse
Affiliation(s)
- Erika Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Emanuela Scarpi
- Biostatistics and Clinical Trials Unit, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giulia Gallerani
- Biosciences Laboratory, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ivan Vannini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Samanta Salvi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Irene Azzali
- Biostatistics and Clinical Trials Unit, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Mattia Melloni
- Biosciences Laboratory, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Salucci
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Michela Battistelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Patrizia Serra
- Biostatistics and Clinical Trials Unit, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Roberta Maltoni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
34
|
Arpino JM, Yin H, Prescott EK, Staples SCR, Nong Z, Li F, Chevalier J, Balint B, O’Neil C, Mortuza R, Milkovich S, Lee JJ, Lorusso D, Sandig M, Hamilton DW, Holdsworth DW, Poepping TL, Ellis CG, Pickering JG. Low-flow intussusception and metastable VEGFR2 signaling launch angiogenesis in ischemic muscle. SCIENCE ADVANCES 2021; 7:eabg9509. [PMID: 34826235 PMCID: PMC8626079 DOI: 10.1126/sciadv.abg9509] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Efforts to promote sprouting angiogenesis in skeletal muscles of individuals with peripheral artery disease have not been clinically successful. We discovered that, contrary to the prevailing view, angiogenesis following ischemic muscle injury in mice was not driven by endothelial sprouting. Instead, real-time imaging revealed the emergence of wide-caliber, primordial conduits with ultralow flow that rapidly transformed into a hierarchical neocirculation by transluminal bridging and intussusception. This process was accelerated by inhibiting vascular endothelial growth factor receptor-2 (VEGFR2). We probed this response by developing the first live-cell model of transluminal endothelial bridging using microfluidics. Endothelial cells subjected to ultralow shear stress could reposition inside the flowing lumen as pillars. Moreover, the low-flow lumen proved to be a privileged location for endothelial cells with reduced VEGFR2 signaling capacity, as VEGFR2 mechanosignals were boosted. These findings redefine regenerative angiogenesis in muscle as an intussusceptive process and uncover a basis for its launch.
Collapse
Affiliation(s)
- John-Michael Arpino
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Hao Yin
- Robarts Research Institute, Western University, London, Canada
| | - Emma K. Prescott
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Sabrina C. R. Staples
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Zengxuan Nong
- Robarts Research Institute, Western University, London, Canada
| | - Fuyan Li
- Robarts Research Institute, Western University, London, Canada
| | - Jacqueline Chevalier
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Brittany Balint
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Caroline O’Neil
- Robarts Research Institute, Western University, London, Canada
| | | | - Stephanie Milkovich
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Jason J. Lee
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medicine, Western University, London, Canada
| | - Daniel Lorusso
- Robarts Research Institute, Western University, London, Canada
| | - Martin Sandig
- Department of Anatomy and Cell Biology, Western University, London, Canada
| | | | - David W. Holdsworth
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Tamie L. Poepping
- Department of Physics and Astronomy, Western University, London, Canada
| | - Christopher G. Ellis
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medicine, Western University, London, Canada
| | - J. Geoffrey Pickering
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medicine, Western University, London, Canada
- Department of Biochemistry, Western University, London, Canada
- Corresponding author.
| |
Collapse
|
35
|
Wang M, Wang K, Liao X, Hu H, Chen L, Meng L, Gao W, Li Q. Carnitine Palmitoyltransferase System: A New Target for Anti-Inflammatory and Anticancer Therapy? Front Pharmacol 2021; 12:760581. [PMID: 34764874 PMCID: PMC8576433 DOI: 10.3389/fphar.2021.760581] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/17/2021] [Indexed: 11/20/2022] Open
Abstract
Lipid metabolism involves multiple biological processes. As one of the most important lipid metabolic pathways, fatty acid oxidation (FAO) and its key rate-limiting enzyme, the carnitine palmitoyltransferase (CPT) system, regulate host immune responses and thus are of great clinical significance. The effect of the CPT system on different tissues or organs is complex: the deficiency or over-activation of CPT disrupts the immune homeostasis by causing energy metabolism disorder and inflammatory oxidative damage and therefore contributes to the development of various acute and chronic inflammatory disorders and cancer. Accordingly, agonists or antagonists targeting the CPT system may become novel approaches for the treatment of diseases. In this review, we first briefly describe the structure, distribution, and physiological action of the CPT system. We then summarize the pathophysiological role of the CPT system in chronic obstructive pulmonary disease, bronchial asthma, acute lung injury, chronic granulomatous disease, nonalcoholic fatty liver disease, hepatic ischemia–reperfusion injury, kidney fibrosis, acute kidney injury, cardiovascular disorders, and cancer. We are also concerned with the current knowledge in either preclinical or clinical studies of various CPT activators/inhibitors for the management of diseases. These compounds range from traditional Chinese medicines to novel nanodevices. Although great efforts have been made in studying the different kinds of CPT agonists/antagonists, only a few pharmaceuticals have been applied for clinical uses. Nevertheless, research on CPT activation or inhibition highlights the pharmacological modulation of CPT-dependent FAO, especially on different CPT isoforms, as a promising anti-inflammatory/antitumor therapeutic strategy for numerous disorders.
Collapse
Affiliation(s)
- Muyun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ximing Liao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiyang Hu
- Department of Vascular Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Liangzhi Chen
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linlin Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Huang H, Li S, Tang Q, Zhu G. Metabolic Reprogramming and Immune Evasion in Nasopharyngeal Carcinoma. Front Immunol 2021; 12:680955. [PMID: 34566954 PMCID: PMC8458828 DOI: 10.3389/fimmu.2021.680955] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/25/2021] [Indexed: 01/31/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor of the nasopharynx mainly characterized by geographic distribution and EBV infection. Metabolic reprogramming, one of the cancer hallmarks, has been frequently reported in NPCs to adapt to internal energy demands and external environmental pressures. Inevitably, the metabolic reprogramming within the tumor cell will lead to a decreased pH value and diverse nutritional supplements in the tumor-infiltrating micro-environment incorporating immune cells, fibroblasts, and endothelial cells. Accumulated evidence indicates that metabolic reprogramming derived from NPC cells may facilitate cancer progression and immunosuppression by cell-cell communications with their surrounding immune cells. This review presents the dysregulated metabolism processes, including glucose, fatty acid, amino acid, nucleotide metabolism, and their mutual interactions in NPC. Moreover, the potential connections between reprogrammed metabolism, tumor immunity, and associated therapy would be discussed in this review. Accordingly, the development of targets on the interactions between metabolic reprogramming and immune cells may provide assistances to overcome the current treatment resistance in NPC patients.
Collapse
Affiliation(s)
- Huimei Huang
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shisheng Li
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qinglai Tang
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Gangcai Zhu
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
37
|
Tura-Ceide O, Smolders VFED, Aventin N, Morén C, Guitart-Mampel M, Blanco I, Piccari L, Osorio J, Rodríguez C, Rigol M, Solanes N, Malandrino A, Kurakula K, Goumans MJ, Quax PHA, Peinado VI, Castellà M, Barberà JA. Derivation and characterisation of endothelial cells from patients with chronic thromboembolic pulmonary hypertension. Sci Rep 2021; 11:18797. [PMID: 34552142 PMCID: PMC8458486 DOI: 10.1038/s41598-021-98320-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/13/2021] [Indexed: 11/09/2022] Open
Abstract
Pulmonary endarterectomy (PEA) resected material offers a unique opportunity to develop an in vitro endothelial cell model of chronic thromboembolic pulmonary hypertension (CTEPH). We aimed to comprehensively analyze the endothelial function, molecular signature, and mitochondrial profile of CTEPH-derived endothelial cells to better understand the pathophysiological mechanisms of endothelial dysfunction behind CTEPH, and to identify potential novel targets for the prevention and treatment of the disease. Isolated cells from specimens obtained at PEA (CTEPH-EC), were characterized based on morphology, phenotype, and functional analyses (in vitro and in vivo tubule formation, proliferation, apoptosis, and migration). Mitochondrial content, morphology, and dynamics, as well as high-resolution respirometry and oxidative stress, were also studied. CTEPH-EC displayed a hyperproliferative phenotype with an increase expression of adhesion molecules and a decreased apoptosis, eNOS activity, migration capacity and reduced angiogenic capacity in vitro and in vivo compared to healthy endothelial cells. CTEPH-EC presented altered mitochondrial dynamics, increased mitochondrial respiration and an unbalanced production of reactive oxygen species and antioxidants. Our study is the foremost comprehensive investigation of CTEPH-EC. Modulation of redox, mitochondrial homeostasis and adhesion molecule overexpression arise as novel targets and biomarkers in CTEPH.
Collapse
Affiliation(s)
- Olga Tura-Ceide
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain. .,Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029, Madrid, Spain. .,Department of Pulmonary Medicine, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institut (IDIBGI), Dr. Josep Trueta University Hospital de Girona, 17190, Girona, Spain.
| | - Valérie F E D Smolders
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain.,Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain.,Department of Vascular Surgery, Leiden University Medical Center, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Núria Aventin
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain
| | - Constanza Morén
- Laboratory of Muscle Research and Mitochondrial Function, Department of Internal Medicine, Hospital Clínic of Barcelona (HCB), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Madrid, Spain
| | - Mariona Guitart-Mampel
- Laboratory of Muscle Research and Mitochondrial Function, Department of Internal Medicine, Hospital Clínic of Barcelona (HCB), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain
| | - Isabel Blanco
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain.,Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029, Madrid, Spain
| | - Lucilla Piccari
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain
| | - Jeisson Osorio
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain.,Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029, Madrid, Spain
| | - Cristina Rodríguez
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain.,Department of Pulmonary Medicine, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institut (IDIBGI), Dr. Josep Trueta University Hospital de Girona, 17190, Girona, Spain
| | - Montserrat Rigol
- Cardiovascular Institute, Hospital Clínic de Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Núria Solanes
- Cardiovascular Institute, Hospital Clínic de Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Andrea Malandrino
- European Molecular Biology Laboratory (EMBL), Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - Kondababu Kurakula
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie Jose Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Victor I Peinado
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain.,Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029, Madrid, Spain
| | - Manuel Castellà
- Department of Cardiovascular Surgery, Cardiovascular Institute, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain. .,Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029, Madrid, Spain.
| |
Collapse
|
38
|
Min J, Zeng T, Roux M, Lazar D, Chen L, Tudzarova S. The Role of HIF1α-PFKFB3 Pathway in Diabetic Retinopathy. J Clin Endocrinol Metab 2021; 106:2505-2519. [PMID: 34019671 PMCID: PMC8372643 DOI: 10.1210/clinem/dgab362] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Indexed: 12/13/2022]
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness for adults in developed countries. Both microvasculopathy and neurodegeneration are implicated in mechanisms of DR development, with neuronal impairment preceding microvascular abnormalities, which is often underappreciated in the clinic. Most current therapeutic strategies, including anti-vascular endothelial growth factor (anti-VEGF)-antibodies, aim at treating the advanced stages (diabetic macular edema and proliferative diabetic retinopathy) and fail to target the neuronal deterioration. Hence, new therapeutic approach(es) intended to address both vascular and neuronal impairment are urgently needed. The hypoxia-inducible factor 1α (HIF1α)-6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) pathway is critically implicated in the islet pathology of diabetes. Recent evidence highlighted the pathway relevance for pathologic angiogenesis and neurodegeneration, two key aspects in DR. PFKFB3 is key to the sprouting angiogenesis, along with VEGF, by determining the endothelial tip-cell competition. Also, PFKFB3-driven glycolysis compromises the antioxidative capacity of neurons leading to neuronal loss and reactive gliosis. Therefore, the HIF1α-PFKFB3 signaling pathway is unique as being a pervasive pathological component across multiple cell types in the retina in the early as well as late stages of DR. A metabolic point-of-intervention based on HIF1α-PFKFB3 targeting thus deserves further consideration in DR.
Collapse
Affiliation(s)
- Jie Min
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Margaretha Roux
- Groote Schuur and Red Cross Children’s Hospital, University of Cape Town, South Africa
| | - David Lazar
- Lazar Retina Ophthalmology, Los Angeles, CA, USA
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Lulu Chen, PhD, Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, Hubei, 430022, China.
| | - Slavica Tudzarova
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Correspondence: Slavica Tudzarova, PhD, Larry Hillblom Islet Research Center, University of California Los Angeles, 10833 Le Conte Ave, CHS 33-165, Los Angeles, CA 90095, USA.
| |
Collapse
|
39
|
Blatchley MR, Hall F, Ntekoumes D, Cho H, Kailash V, Vazquez‐Duhalt R, Gerecht S. Discretizing Three-Dimensional Oxygen Gradients to Modulate and Investigate Cellular Processes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100190. [PMID: 34151527 PMCID: PMC8292886 DOI: 10.1002/advs.202100190] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 05/05/2021] [Indexed: 05/26/2023]
Abstract
With the increased realization of the effect of oxygen (O2 ) deprivation (hypoxia) on cellular processes, recent efforts have focused on the development of engineered systems to control O2 concentrations and establish biomimetic O2 gradients to study and manipulate cellular behavior. Nonetheless, O2 gradients present in 3D engineered platforms result in diverse cell behavior across the O2 gradient, making it difficult to identify and study O2 sensitive signaling pathways. Using a layer-by-layer assembled O2 -controllable hydrogel, the authors precisely control O2 concentrations and study uniform cell behavior in discretized O2 gradients, then recapitulate the dynamics of cluster-based vasculogenesis, one mechanism for neovessel formation, and show distinctive gene expression patterns remarkably correlate to O2 concentrations. Using RNA sequencing, it is found that time-dependent regulation of cyclic adenosine monophosphate signaling enables cell survival and clustering in the high stress microenvironments. Various extracellular matrix modulators orchestrate hypoxia-driven endothelial cell clustering. Finally, clustering is facilitated by regulators of cell-cell interactions, mainly vascular cell adhesion molecule 1. Taken together, novel regulators of hypoxic cluster-based vasculogenesis are identified, and evidence for the utility of a unique platform is provided to study dynamic cellular responses to 3D hypoxic environments, with broad applicability in development, regeneration, and disease.
Collapse
Affiliation(s)
- Michael R. Blatchley
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of Chemical and Biomolecular EngineeringInstitute for NanoBioTechnology and Johns Hopkins Physical Sciences‐Oncology CenterJohns Hopkins UniversityBaltimoreMD21218USA
| | - Franklyn Hall
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of Chemical and Biomolecular EngineeringInstitute for NanoBioTechnology and Johns Hopkins Physical Sciences‐Oncology CenterJohns Hopkins UniversityBaltimoreMD21218USA
| | - Dimitris Ntekoumes
- Department of Chemical and Biomolecular EngineeringInstitute for NanoBioTechnology and Johns Hopkins Physical Sciences‐Oncology CenterJohns Hopkins UniversityBaltimoreMD21218USA
| | - Hyunwoo Cho
- Department of Chemical and Biomolecular EngineeringInstitute for NanoBioTechnology and Johns Hopkins Physical Sciences‐Oncology CenterJohns Hopkins UniversityBaltimoreMD21218USA
| | - Vidur Kailash
- Department of BiophysicsJohns Hopkins UniversityBaltimoreMD21218USA
| | - Rafael Vazquez‐Duhalt
- Department of BionanotechnologyCenter for Nanosciences and NanotechnologyNational Autonomous University of MexicoEnsenadaBaja California22800Mexico
| | - Sharon Gerecht
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of Chemical and Biomolecular EngineeringInstitute for NanoBioTechnology and Johns Hopkins Physical Sciences‐Oncology CenterJohns Hopkins UniversityBaltimoreMD21218USA
- Department of Materials Science and EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of OncologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
| |
Collapse
|
40
|
Blin JA, Hamid RA, Khaza'ai H. Bioactive fractions and compound of Ardisia crispa roots exhibit anti-arthritic properties mediated via angiogenesis inhibition in vitro. BMC Complement Med Ther 2021; 21:176. [PMID: 34172047 PMCID: PMC8235828 DOI: 10.1186/s12906-021-03341-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/03/2021] [Indexed: 01/18/2023] Open
Abstract
Background Ardisia crispa (Thunb.) A.DC (Primulaceae), is a medicinal herb traditionally used by Asian people as remedies to cure inflammatory related diseases, including rheumatism. The plant roots possess various pharmacological activities including antipyretic, anti-inflammation and antitumor. Previous phytochemical studies of the plant roots have identified long chain alkyl-1,4-benzoquinones as major constituents, together with other phytochemicals. Hexane fraction of the plant roots (ACRH), was previously reported with anti-angiogenic and anti-arthritic properties, while its effect on their anti-arthritic in vitro, is yet unrevealed. Considering the significance of angiogenesis inhibition in developing new anti-arthritic agent, thus we investigated the anti-arthritic potential of Ardisia crispa roots by suppressing angiogenesis, in vitro. Methods Ardisia crispa roots hexane extract (ACRH) was prepared from the plant roots using absolute n-hexane. ACRH was fractionated into quinone-rich fraction (QRF) and further isolated to yield benzoquinonoid compound (BQ), respectively. In vitro experiments using VEGF-induced human umbilical vein endothelial cells (HUVECs) and IL-1β-induced human fibroblast-like synoviocytes for rheumatoid arthritis (HFLS-RA) were performed to evaluate the effects of these samples on VEGF-induced HUVECs proliferation and tube formation, and towards IL-1β-induced HFLS-RA proliferation, invasion, and apoptosis, respectively. Therapeutic concentrations (0.05, 0.5, and 5 μg/mL) tested in this study were predetermined based on the IC50 values obtained from the MTT assay. Results ACRH, QRF, and BQ exerted concentration-independent antiproliferative effects on VEGF-induced HUVECs and IL-1β-induced HFLS-RA, with IC50 values at 1.09 ± 0.18, 3.85 ± 0.26, and 1.34 ± 0.16 μg/mL in HUVECs; and 3.60 ± 1.38, 4.47 ± 0.34, and 1.09 ± 0.09 μg/mL in HFLS-RA, respectively. Anti-angiogenic properties of these samples were verified via significant inhibition on VEGF-induced HUVECs tube formation, in a concentration-independent manner. The invasiveness of IL-1β-induced HFLS-RA was also significantly inhibited in a concentration-independent manner by all samples. ACRH and BQ, but not QRF, significantly enhanced the apoptosis of IL-1β-induced HFLS-RA elicited at their highest concentration (5 μg/mL) (P < 0.05). Conclusions These findings highlight the bioactive fractions and compound from Ardisia crispa roots as potential anti-arthritic agents by inhibiting both HUVECs and HFLS-RA’s cellular functions in vitro, possibly mediated via their anti-angiogenic effects. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03341-y.
Collapse
Affiliation(s)
- Joan Anak Blin
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Roslida Abdul Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Huzwah Khaza'ai
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| |
Collapse
|
41
|
Zheng R, Li F, Li F, Gong A. Targeting tumor vascularization: promising strategies for vascular normalization. J Cancer Res Clin Oncol 2021; 147:2489-2505. [PMID: 34148156 DOI: 10.1007/s00432-021-03701-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022]
Abstract
Tumor recurrence after the clinical cure of tumor often results from the presence of an abnormal microenvironment, including an aberrant vasculature. The tumor microenvironment is rich in pro-angiogenic factors but lacks pro-maturation factors. Pro-angiogenic conditions in the tumor microenvironment, such as hypoxia, are double-edged swords, promoting both the repair of normal tissues and the development of an abnormal blood vessel network. The coexistence of perfusion and hypoxic zones and uneven blood vessel distribution in tumor tissues profoundly influence tumor deterioration, recurrence, and metastasis. Traditional anti-angiogenic therapies have shown limited efficacy, and promote drug resistance, and even metastasis. In contrast, vascular normalization therapy induces a more physiological-like state, leading to better outcomes and fewer side effects. Vascular normalization entails modifying the tumor vascular system to improve tumor oxygenation and substance transport, thereby contributing to improving the efficacy of radiotherapy, chemotherapy, and immunotherapy. This review mainly focuses on the process of tumor vascularization; potential therapeutic targets, including cells, metabolism, signaling pathways, and angiogenesis-related genes; and possible strategies to normalize blood vessels through regulating tumor vessel generation, the development of tumor vessels, and blood vessel fusion and pruning.
Collapse
Affiliation(s)
- Ruiqi Zheng
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China
| | - Feifan Li
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China
| | - Fengcen Li
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China
| | - Aihua Gong
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China.
| |
Collapse
|
42
|
Ushio-Fukai M, Ash D, Nagarkoti S, Belin de Chantemèle EJ, Fulton DJR, Fukai T. Interplay Between Reactive Oxygen/Reactive Nitrogen Species and Metabolism in Vascular Biology and Disease. Antioxid Redox Signal 2021; 34:1319-1354. [PMID: 33899493 PMCID: PMC8418449 DOI: 10.1089/ars.2020.8161] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS; e.g., superoxide [O2•-] and hydrogen peroxide [H2O2]) and reactive nitrogen species (RNS; e.g., nitric oxide [NO•]) at the physiological level function as signaling molecules that mediate many biological responses, including cell proliferation, migration, differentiation, and gene expression. By contrast, excess ROS/RNS, a consequence of dysregulated redox homeostasis, is a hallmark of cardiovascular disease. Accumulating evidence suggests that both ROS and RNS regulate various metabolic pathways and enzymes. Recent studies indicate that cells have mechanisms that fine-tune ROS/RNS levels by tight regulation of metabolic pathways, such as glycolysis and oxidative phosphorylation. The ROS/RNS-mediated inhibition of glycolytic pathways promotes metabolic reprogramming away from glycolytic flux toward the oxidative pentose phosphate pathway to generate nicotinamide adenine dinucleotide phosphate (NADPH) for antioxidant defense. This review summarizes our current knowledge of the mechanisms by which ROS/RNS regulate metabolic enzymes and cellular metabolism and how cellular metabolism influences redox homeostasis and the pathogenesis of disease. A full understanding of these mechanisms will be important for the development of new therapeutic strategies to treat diseases associated with dysregulated redox homeostasis and metabolism. Antioxid. Redox Signal. 34, 1319-1354.
Collapse
Affiliation(s)
- Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Dipankar Ash
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Sheela Nagarkoti
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - David J R Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| |
Collapse
|
43
|
Yakubov E, Eibl T, Hammer A, Holtmannspötter M, Savaskan N, Steiner HH. Therapeutic Potential of Selenium in Glioblastoma. Front Neurosci 2021; 15:666679. [PMID: 34121995 PMCID: PMC8194316 DOI: 10.3389/fnins.2021.666679] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/04/2021] [Indexed: 01/06/2023] Open
Abstract
Little progress has been made in the long-term management of malignant brain tumors, leaving patients with glioblastoma, unfortunately, with a fatal prognosis. Glioblastoma remains the most aggressive primary brain cancer in adults. Similar to other cancers, glioblastoma undergoes a cellular metabolic reprogramming to form an oxidative tumor microenvironment, thereby fostering proliferation, angiogenesis and tumor cell survival. Latest investigations revealed that micronutrients, such as selenium, may have positive effects in glioblastoma treatment, providing promising chances regarding the current limitations in surgical treatment and radiochemotherapy outcomes. Selenium is an essential micronutrient with anti-oxidative and anti-cancer properties. There is additional evidence of Se deficiency in patients suffering from brain malignancies, which increases its importance as a therapeutic option for glioblastoma therapy. It is well known that selenium, through selenoproteins, modulates metabolic pathways and regulates redox homeostasis. Therefore, selenium impacts on the interaction in the tumor microenvironment between tumor cells, tumor-associated cells and immune cells. In this review we take a closer look at the current knowledge about the potential of selenium on glioblastoma, by focusing on brain edema, glioma-related angiogenesis, and cells in tumor microenvironment such as glioma-associated microglia/macrophages.
Collapse
Affiliation(s)
- Eduard Yakubov
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg, Germany
| | - Thomas Eibl
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg, Germany
| | - Alexander Hammer
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg, Germany
| | | | - Nicolai Savaskan
- Department of Neurosurgery, University Medical School Hospital, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,BiMECON Ent., Berlin, Germany
| | | |
Collapse
|
44
|
Şen Ö, Emanet M, Ciofani G. Nanotechnology-Based Strategies to Evaluate and Counteract Cancer Metastasis and Neoangiogenesis. Adv Healthc Mater 2021; 10:e2002163. [PMID: 33763992 PMCID: PMC7610913 DOI: 10.1002/adhm.202002163] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/11/2021] [Indexed: 12/15/2022]
Abstract
Cancer metastasis is the major cause of cancer-related morbidity and mortality. It represents one of the greatest challenges in cancer therapy, both because of the ability of metastatic cells to spread into different organs, and because of the consequent heterogeneity that characterizes primary and metastatic tumors. Nanomaterials can potentially be used as targeting or detection agents owing to unique chemical and physical features that allow tailored and tunable theranostic functions. This review highlights nanomaterial-based approaches in the detection and treatment of cancer metastasis, with a special focus on the evaluation of nanostructure effects on cell migration, invasion, and angiogenesis in the tumor microenvironment.
Collapse
Affiliation(s)
- Özlem Şen
- Istituto Italiano di TecnologiaSmart Bio‐InterfacesViale Rinaldo Piaggio 34PontederaPisa56025Italy
| | - Melis Emanet
- Istituto Italiano di TecnologiaSmart Bio‐InterfacesViale Rinaldo Piaggio 34PontederaPisa56025Italy
- Sabanci University Nanotechnology Research and Application Center (SUNUM)Sabanci UniversityUniversite Caddesi 27‐1TuzlaIstanbul34956Turkey
| | - Gianni Ciofani
- Istituto Italiano di TecnologiaSmart Bio‐InterfacesViale Rinaldo Piaggio 34PontederaPisa56025Italy
| |
Collapse
|
45
|
Salmina AB, Kharitonova EV, Gorina YV, Teplyashina EA, Malinovskaya NA, Khilazheva ED, Mosyagina AI, Morgun AV, Shuvaev AN, Salmin VV, Lopatina OL, Komleva YK. Blood-Brain Barrier and Neurovascular Unit In Vitro Models for Studying Mitochondria-Driven Molecular Mechanisms of Neurodegeneration. Int J Mol Sci 2021; 22:4661. [PMID: 33925080 PMCID: PMC8125678 DOI: 10.3390/ijms22094661] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
Pathophysiology of chronic neurodegeneration is mainly based on complex mechanisms related to aberrant signal transduction, excitation/inhibition imbalance, excitotoxicity, synaptic dysfunction, oxidative stress, proteotoxicity and protein misfolding, local insulin resistance and metabolic dysfunction, excessive cell death, development of glia-supported neuroinflammation, and failure of neurogenesis. These mechanisms tightly associate with dramatic alterations in the structure and activity of the neurovascular unit (NVU) and the blood-brain barrier (BBB). NVU is an ensemble of brain cells (brain microvessel endothelial cells (BMECs), astrocytes, pericytes, neurons, and microglia) serving for the adjustment of cell-to-cell interactions, metabolic coupling, local microcirculation, and neuronal excitability to the actual needs of the brain. The part of the NVU known as a BBB controls selective access of endogenous and exogenous molecules to the brain tissue and efflux of metabolites to the blood, thereby providing maintenance of brain chemical homeostasis critical for efficient signal transduction and brain plasticity. In Alzheimer's disease, mitochondria are the target organelles for amyloid-induced neurodegeneration and alterations in NVU metabolic coupling or BBB breakdown. In this review we discuss understandings on mitochondria-driven NVU and BBB dysfunction, and how it might be studied in current and prospective NVU/BBB in vitro models for finding new approaches for the efficient pharmacotherapy of Alzheimer's disease.
Collapse
Affiliation(s)
- Alla B. Salmina
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
- Research Center of Neurology, 125367 Moscow, Russia
| | - Ekaterina V. Kharitonova
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Yana V. Gorina
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Elena A. Teplyashina
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Natalia A. Malinovskaya
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Elena D. Khilazheva
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Angelina I. Mosyagina
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Andrey V. Morgun
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Anton N. Shuvaev
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Vladimir V. Salmin
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Olga L. Lopatina
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| | - Yulia K. Komleva
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (E.V.K.); (Y.V.G.); (E.A.T.); (N.A.M.); (E.D.K.); (A.I.M.); (A.V.M.); (A.N.S.); (V.V.S.); (O.L.L.); (Y.K.K.)
| |
Collapse
|
46
|
Chen X, Zhang M, Chen L, Zhou Z, Chen B, Wang C, Xie Y, Zhang Y. Roxarsone Promotes Glycolysis and Angiogenesis by Inducing Hypoxia-Inducible Factor-1α In Vitro and In Vivo. ACS OMEGA 2021; 6:9559-9566. [PMID: 33869936 PMCID: PMC8047655 DOI: 10.1021/acsomega.1c00072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/18/2021] [Indexed: 06/12/2023]
Abstract
Roxarsone (Rox) is an organic arsenic compound used as a feed additive to promote animal growth. The release of Rox into the environment poses risks to human health. Rox demonstrated tumor-promoting and proangiogenic effects in xenograft models. Increasing studies revealed the tight relationship among angiogenesis, carcinogenesis, tumorigenesis, and glycolysis. Glycolysis, via hypoxia-inducible factor-1α (HIF-1α), controls vascular endothelial cell (VEC) growth. To date, there has been no literature report on the effect of Rox on HIF-1α-dependent glycolysis. Herein, we report that Rox promoted glycolysis in rat VECs, as shown by the increased adenosine triphosphate production, the lactic acid release, the activity and content of aldolase (ALD), and the expression levels of ALD A and glucose transporter 1 (GLUT1). Rox also increased the cellular levels of HIF-1α. Treatment with the HIF-1α inhibitor YC-1 reversed Rox-increased ALD A and GLUT1 levels and attenuated Rox-induced VEC viability, suggesting that Rox-induced HIF-1α contributes to the glycolytic and angiogenic effects of Rox. Rox also promoted tumor growth and angiogenesis and increased the levels of ALD A, GLUT1, and HIF-1α in the tumor tissue of a mouse xenograft model, whereas these effects were abolished using YC-1. Our findings indicated that Rox induces HIF-1α in VECs to promote glycolysis and angiogenesis thus enhancing the tumor growth.
Collapse
Affiliation(s)
- Xin Chen
- Department
of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu, China
- Joint
International Research Laboratory of Agriculture and Agri-Product
Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009 Jiangsu, China
| | - Meng Zhang
- Department
of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu, China
| | - Linzhongri Chen
- Department
of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu, China
| | - Zhiqiang Zhou
- Department
of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu, China
| | - Binlin Chen
- Department
of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu, China
| | - Cunkai Wang
- Department
of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu, China
| | - Yang Xie
- Department
of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu, China
| | - Yumei Zhang
- Department
of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu, China
- Joint
International Research Laboratory of Agriculture and Agri-Product
Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009 Jiangsu, China
| |
Collapse
|
47
|
Baird A, Oelsner L, Fisher C, Witte M, Huynh M. A multiscale computational model of angiogenesis after traumatic brain injury, investigating the role location plays in volumetric recovery. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:3227-3257. [PMID: 34198383 DOI: 10.3934/mbe.2021161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Vascular endothelial growth factor (VEGF) is a key protein involved in the process of angiogenesis. VEGF is of particular interest after a traumatic brain injury (TBI), as it re-establishes the cerebral vascular network in effort to allow for proper cerebral blood flow and thereby oxygenation of damaged brain tissue. For this reason, angiogenesis is critical in the progression and recovery of TBI patients in the days and weeks post injury. Although well established experimental work has led to advances in our understanding of TBI and the progression of angiogenisis, many constraints still exist with existing methods, especially when considering patient progression in the days following injury. To better understand the healing process on the proposed time scales, we develop a computational model that quickly simulates vessel growth and recovery by coupling VEGF and its interactions with its associated receptors to a physiologically inspired fractal model of the microvascular re-growth. We use this model to clarify the role that diffusivity, receptor kinetics and location of the TBI play in overall blood volume restoration in the weeks post injury and show that proper therapeutic angiogenesis, or vasculogenic therapies, could speed recovery of the patient as a function of the location of injury.
Collapse
Affiliation(s)
- Austin Baird
- Applied Research Associates Inc., Advanced Modeling & Simulation Systems Directorate, 8537 Six Forks Rd, Raleigh, NC 27615, USA
| | - Laura Oelsner
- Varian Medical Systems, 3100 Hansen Way, Palo Alto, CA 94304, USA
| | - Charles Fisher
- Applied Research Associates Inc., Advanced Modeling & Simulation Systems Directorate, 8537 Six Forks Rd, Raleigh, NC 27615, USA
| | - Matt Witte
- Applied Research Associates Inc., Advanced Modeling & Simulation Systems Directorate, 8537 Six Forks Rd, Raleigh, NC 27615, USA
| | - My Huynh
- Applied Research Associates Inc., Advanced Modeling & Simulation Systems Directorate, 8537 Six Forks Rd, Raleigh, NC 27615, USA
| |
Collapse
|
48
|
Luca AC, Miron IC, Trandafir LM, Cojocaru E, Pădureţ IA, Trandafirescu MF, Iordache AC, Ţarcă E. Morphological, genetic and clinical correlations in infantile hemangiomas and their mimics. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2021; 61:687-695. [PMID: 33817710 PMCID: PMC8112746 DOI: 10.47162/rjme.61.3.07] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Infantile hemangiomas (IHs) are the most frequent pediatric benign vascular tumors, with a reported incidence of 5% to 10%. They have self-limiting evolution pattern divided into a growth phase in the first 12 months and a regression one, that may take up to 10 years. Occasionally, hemangiomas might lead to local or systemic complications, depending on their morphological characteristics. The first line of treatment is β-blockers, such as Propranolol, Timolol, Nadolol, administered either locally or systemically. Newer therapeutic strategies involving laser therapy and angiotensin-converting enzyme inhibitors are being studied, while older treatment modalities like corticosteroids, Imiquimod, Vincristine, Bleomycin and Interferon-α have become second line therapy options. Before establishing the appropriate treatment, clinical, histological, and imaging investigations are required.
Collapse
Affiliation(s)
- Alina Costina Luca
- Department of Morphofunctional Sciences I - Pathology, Department of Mother and Child Medicine - Pediatrics, Grigore T. Popa University of Medicine and Pharmacy, Iaşi, Romania; ,
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Popovic N, Hooker E, Barabino A, Flamier A, Provost F, Buscarlet M, Bernier G, Larrivée B. COCO/DAND5 inhibits developmental and pathological ocular angiogenesis. EMBO Mol Med 2021; 13:e12005. [PMID: 33587337 PMCID: PMC7933934 DOI: 10.15252/emmm.202012005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
Neovascularization contributes to multiple visual disorders including age-related macular degeneration (AMD) and retinopathy of prematurity. Current therapies for treating ocular angiogenesis are centered on the inhibition of vascular endothelial growth factor (VEGF). While clinically effective, some AMD patients are refractory or develop resistance to anti-VEGF therapies and concerns of increased risks of developing geographic atrophy following long-term treatment have been raised. Identification of alternative pathways to inhibit pathological angiogenesis is thus important. We have identified a novel inhibitor of angiogenesis, COCO, a member of the Cerberus-related DAN protein family. We demonstrate that COCO inhibits sprouting, migration and cellular proliferation of cultured endothelial cells. Intravitreal injections of COCO inhibited retinal vascularization during development and in models of retinopathy of prematurity. COCO equally abrogated angiogenesis in models of choroidal neovascularization. Mechanistically, COCO inhibited TGFβ and BMP pathways and altered energy metabolism and redox balance of endothelial cells. Together, these data show that COCO is an inhibitor of retinal and choroidal angiogenesis, possibly representing a therapeutic option for the treatment of neovascular ocular diseases.
Collapse
Affiliation(s)
- Natalija Popovic
- Faculty of MedicineUniversity of MontrealMontrealQCCanada
- Hôpital Maisonneuve Rosemont Research CentreMontrealQCCanada
| | - Erika Hooker
- Faculty of MedicineUniversity of MontrealMontrealQCCanada
- Hôpital Maisonneuve Rosemont Research CentreMontrealQCCanada
| | - Andrea Barabino
- Hôpital Maisonneuve Rosemont Research CentreMontrealQCCanada
- Department of NeurosciencesUniversity of MontrealMontrealQCCanada
| | - Anthony Flamier
- Hôpital Maisonneuve Rosemont Research CentreMontrealQCCanada
- Department of NeurosciencesUniversity of MontrealMontrealQCCanada
- Present address:
Whitehead Institute of Biomedical ResearchCambridgeMAUSA
| | | | | | - Gilbert Bernier
- Faculty of MedicineUniversity of MontrealMontrealQCCanada
- Hôpital Maisonneuve Rosemont Research CentreMontrealQCCanada
- Department of NeurosciencesUniversity of MontrealMontrealQCCanada
| | - Bruno Larrivée
- Faculty of MedicineUniversity of MontrealMontrealQCCanada
- Hôpital Maisonneuve Rosemont Research CentreMontrealQCCanada
- Department of OphthalmologyUniversity of MontrealMontrealQCCanada
| |
Collapse
|
50
|
Luxán G, Dimmeler S. The vasculature: a therapeutic target in heart failure? Cardiovasc Res 2021; 118:53-64. [PMID: 33620071 PMCID: PMC8752358 DOI: 10.1093/cvr/cvab047] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
It is well established that the vasculature plays a crucial role in maintaining oxygen and nutrients supply to the heart. Increasing evidence further suggest that the microcirculation has additional roles in supporting a healthy microenvironment. Heart failure is well known to be associated with changes and functional impairment of the microvasculature. The specific ablation of protective signals in endothelial cells in experimental models is sufficient to induce heart failure. Therefore, restoring a healthy endothelium and microcirculation may be a valuable therapeutic strategy to treat heart failure. The present review article will summarize the current understanding of the vascular contribution to heart failure with reduced or preserved ejection fraction. Novel therapeutic approaches including next generation pro-angiogenic therapies and non-coding RNA therapeutics, as well as the targeting of metabolites or metabolic signaling, vascular inflammation and senescence will be discussed.
Collapse
Affiliation(s)
- Guillermo Luxán
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany, German Center for Cardiovascular Research DZHK, Berlin, Germany, partner site Frankfurt Rhine-Main, Germany, Cardiopulmonary Institute, Goethe University Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany, German Center for Cardiovascular Research DZHK, Berlin, Germany, partner site Frankfurt Rhine-Main, Germany, Cardiopulmonary Institute, Goethe University Frankfurt, Germany
| |
Collapse
|