1
|
Kuintzle R, Santat LA, Elowitz MB. Diversity in Notch ligand-receptor signaling interactions. eLife 2025; 12:RP91422. [PMID: 39751380 PMCID: PMC11698495 DOI: 10.7554/elife.91422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
The Notch signaling pathway uses families of ligands and receptors to transmit signals to nearby cells. These components are expressed in diverse combinations in different cell types, interact in a many-to-many fashion, both within the same cell (in cis) and between cells (in trans), and their interactions are modulated by Fringe glycosyltransferases. A fundamental question is how the strength of Notch signaling depends on which pathway components are expressed, at what levels, and in which cells. Here, we used a quantitative, bottom-up, cell-based approach to systematically characterize trans-activation, cis-inhibition, and cis-activation signaling efficiencies across a range of ligand and Fringe expression levels in Chinese hamster and mouse cell lines. Each ligand (Dll1, Dll4, Jag1, and Jag2) and receptor variant (Notch1 and Notch2) analyzed here exhibited a unique profile of interactions, Fringe dependence, and signaling outcomes. All four ligands were able to bind receptors in cis and in trans, and all ligands trans-activated both receptors, although Jag1-Notch1 signaling was substantially weaker than other ligand-receptor combinations. Cis-interactions were predominantly inhibitory, with the exception of the Dll1- and Dll4-Notch2 pairs, which exhibited cis-activation stronger than trans-activation. Lfng strengthened Delta-mediated trans-activation and weakened Jagged-mediated trans-activation for both receptors. Finally, cis-ligands showed diverse cis-inhibition strengths, which depended on the identity of the trans-ligand as well as the receptor. The map of receptor-ligand-Fringe interaction outcomes revealed here should help guide rational perturbation and control of the Notch pathway.
Collapse
Affiliation(s)
- Rachael Kuintzle
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Leah A Santat
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
- Howard Hughes Medical Institute, California Institute of TechnologyPasadenaUnited States
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
- Howard Hughes Medical Institute, California Institute of TechnologyPasadenaUnited States
| |
Collapse
|
2
|
Mora P, Laisné M, Bourguignon C, Rouault P, Jaspard-Vinassa B, Maître M, Gadeau AP, Renault MA, Horng S, Couffinhal T, Chapouly C. Astrocytic DLL4-NOTCH1 signaling pathway promotes neuroinflammation via the IL-6-STAT3 axis. J Neuroinflammation 2024; 21:258. [PMID: 39390606 PMCID: PMC11468415 DOI: 10.1186/s12974-024-03246-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
Under neuroinflammatory conditions, astrocytes acquire a reactive phenotype that drives acute inflammatory injury as well as chronic neurodegeneration. We hypothesized that astrocytic Delta-like 4 (DLL4) may interact with its receptor NOTCH1 on neighboring astrocytes to regulate astrocyte reactivity via downstream juxtacrine signaling pathways. Here we investigated the role of astrocytic DLL4 on neurovascular unit homeostasis under neuroinflammatory conditions. We probed for downstream effectors of the DLL4-NOTCH1 axis and targeted these for therapy in two models of CNS inflammatory disease. We first demonstrated that astrocytic DLL4 is upregulated during neuroinflammation, both in mice and humans, driving astrocyte reactivity and subsequent blood-brain barrier permeability and inflammatory infiltration. We then showed that the DLL4-mediated NOTCH1 signaling in astrocytes directly drives IL-6 levels, induces STAT3 phosphorylation promoting upregulation of astrocyte reactivity markers, pro-permeability factor secretion and consequent blood-brain barrier destabilization. Finally we revealed that blocking DLL4 with antibodies improves experimental autoimmune encephalomyelitis symptoms in mice, identifying a potential novel therapeutic strategy for CNS autoimmune demyelinating disease. As a general conclusion, this study demonstrates that DLL4-NOTCH1 signaling is not only a key pathway in vascular development and angiogenesis, but also in the control of astrocyte reactivity during neuroinflammation.
Collapse
Affiliation(s)
- Pierre Mora
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Margaux Laisné
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Célia Bourguignon
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Paul Rouault
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Béatrice Jaspard-Vinassa
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Marlène Maître
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, F-33000, France
| | - Alain-Pierre Gadeau
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Marie-Ange Renault
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Sam Horng
- Department of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Thierry Couffinhal
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Candice Chapouly
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France.
| |
Collapse
|
3
|
Guelfi S, Hodivala-Dilke K, Bergers G. Targeting the tumour vasculature: from vessel destruction to promotion. Nat Rev Cancer 2024; 24:655-675. [PMID: 39210063 DOI: 10.1038/s41568-024-00736-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
As angiogenesis was recognized as a core hallmark of cancer growth and survival, several strategies have been implemented to target the tumour vasculature. Yet to date, attempts have rarely been so diverse, ranging from vessel growth inhibition and destruction to vessel normalization, reprogramming and vessel growth promotion. Some of these strategies, combined with standard of care, have translated into improved cancer therapies, but their successes are constrained to certain cancer types. This Review provides an overview of these vascular targeting approaches and puts them into context based on our subsequent improved understanding of the tumour vasculature as an integral part of the tumour microenvironment with which it is functionally interlinked. This new knowledge has already led to dual targeting of the vascular and immune cell compartments and sets the scene for future investigations of possible alternative approaches that consider the vascular link with other tumour microenvironment components for improved cancer therapy.
Collapse
Affiliation(s)
- Sophie Guelfi
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology and KU Leuven, Leuven, Belgium
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, UK.
| | - Gabriele Bergers
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology and KU Leuven, Leuven, Belgium.
| |
Collapse
|
4
|
Zhang S, Yu M, Li M, He M, Xie L, Huo F, Tian W. Notch Signaling Hydrogels Enable Rapid Vascularization and Promote Dental Pulp Tissue Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310285. [PMID: 39013081 PMCID: PMC11425206 DOI: 10.1002/advs.202310285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/07/2024] [Indexed: 07/18/2024]
Abstract
Successful dental pulp regeneration is closely associated with rapid revascularization and angiogenesis, processes driven by the Jagged1(JAG1)/Notch signaling pathway. However, soluble Notch ligands have proven ineffective in activating this pathway. To overcome this limitation, a Notch signaling hydrogel is developed by indirectly immobilizing JAG1, aimed at precisely directing the regeneration of vascularized pulp tissue. This hydrogel displays favorable mechanical properties and biocompatibility. Cultivating dental pulp stem cells (DPSCs) and endothelial cells (ECs) on this hydrogel significantly upregulate Notch target genes and key proangiogenic markers expression. Three-dimensional (3D) culture assays demonstrate Notch signaling hydrogels improve effectiveness by facilitating encapsulated cell differentiation, enhancing their paracrine functions, and promoting capillary lumen formation. Furthermore, it effectively communicates with the Wnt signaling pathway, creating an odontoinductive microenvironment for pulp-dentin complex formation. In vivo studies show that short-term transplantation of the Notch signaling hydrogel accelerates angiogenesis, stabilizes capillary-like structures, and improves cell survival. Long-term transplantation further confirms its capability to promote the formation of pulp-like tissues rich in blood vessels and peripheral nerve-like structures. In conclusion, this study introduces a feasible and effective hydrogel tailored to specifically regulate the JAG1/Notch signaling pathway, showing potential in advancing regenerative strategies for dental pulp tissue.
Collapse
Affiliation(s)
- Siyuan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Mei Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Maojiao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Min He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Li Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Fangjun Huo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
5
|
Kuintzle R, Santat LA, Elowitz MB. Diversity in Notch ligand-receptor signaling interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.24.554677. [PMID: 37662208 PMCID: PMC10473737 DOI: 10.1101/2023.08.24.554677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The Notch signaling pathway uses families of ligands and receptors to transmit signals to nearby cells. These components are expressed in diverse combinations in different cell types, interact in a many-to-many fashion, both within the same cell (in cis) and between cells (in trans), and their interactions are modulated by Fringe glycosyltransferases. A fundamental question is how the strength of Notch signaling depends on which pathway components are expressed, at what levels, and in which cells. Here, we used a quantitative, bottom-up, cell-based approach to systematically characterize trans-activation, cis-inhibition, and cis-activation signaling efficiencies across a range of ligand and Fringe expression levels in two mammalian cell types. Each ligand (Dll1, Dll4, Jag1, and Jag2) and receptor variant (Notch1 and Notch2) analyzed here exhibited a unique profile of interactions, Fringe-dependence, and signaling outcomes. All four ligands were able to bind receptors in cis and in trans, and all ligands trans-activated both receptors, although Jag1-Notch1 signaling was substantially weaker than other ligand-receptor combinations. Cis-interactions were predominantly inhibitory, with the exception of the Dll1- and Dll4-Notch2 pairs, which exhibited cis-activation stronger than trans-activation. Lfng strengthened Delta-mediated trans-activation and weakened Jagged-mediated trans-activation for both receptors. Finally, cis-ligands showed diverse cis-inhibition strengths, which depended on the identity of the trans-ligand as well as the receptor. The map of receptor-ligand-Fringe interaction outcomes revealed here should help guide rational perturbation and control of the Notch pathway.
Collapse
|
6
|
Shi Q, Xue C, Zeng Y, Yuan X, Chu Q, Jiang S, Wang J, Zhang Y, Zhu D, Li L. Notch signaling pathway in cancer: from mechanistic insights to targeted therapies. Signal Transduct Target Ther 2024; 9:128. [PMID: 38797752 PMCID: PMC11128457 DOI: 10.1038/s41392-024-01828-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/31/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Notch signaling, renowned for its role in regulating cell fate, organ development, and tissue homeostasis across metazoans, is highly conserved throughout evolution. The Notch receptor and its ligands are transmembrane proteins containing epidermal growth factor-like repeat sequences, typically necessitating receptor-ligand interaction to initiate classical Notch signaling transduction. Accumulating evidence indicates that the Notch signaling pathway serves as both an oncogenic factor and a tumor suppressor in various cancer types. Dysregulation of this pathway promotes epithelial-mesenchymal transition and angiogenesis in malignancies, closely linked to cancer proliferation, invasion, and metastasis. Furthermore, the Notch signaling pathway contributes to maintaining stem-like properties in cancer cells, thereby enhancing cancer invasiveness. The regulatory role of the Notch signaling pathway in cancer metabolic reprogramming and the tumor microenvironment suggests its pivotal involvement in balancing oncogenic and tumor suppressive effects. Moreover, the Notch signaling pathway is implicated in conferring chemoresistance to tumor cells. Therefore, a comprehensive understanding of these biological processes is crucial for developing innovative therapeutic strategies targeting Notch signaling. This review focuses on the research progress of the Notch signaling pathway in cancers, providing in-depth insights into the potential mechanisms of Notch signaling regulation in the occurrence and progression of cancer. Additionally, the review summarizes pharmaceutical clinical trials targeting Notch signaling for cancer therapy, aiming to offer new insights into therapeutic strategies for human malignancies.
Collapse
Affiliation(s)
- Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shuwen Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jinzhi Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yaqi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
7
|
Mandula JK, Sierra-Mondragon RA, Jimenez RV, Chang D, Mohamed E, Chang S, Vazquez-Martinez JA, Cao Y, Anadon CM, Lee SB, Das S, Rocha-Munguba L, Pham VM, Li R, Tarhini AA, Furqan M, Dalton W, Churchman M, Moran-Segura CM, Nguyen J, Perez B, Kojetin DJ, Obermayer A, Yu X, Chen A, Shaw TI, Conejo-Garcia JR, Rodriguez PC. Jagged2 targeting in lung cancer activates anti-tumor immunity via Notch-induced functional reprogramming of tumor-associated macrophages. Immunity 2024; 57:1124-1140.e9. [PMID: 38636522 PMCID: PMC11096038 DOI: 10.1016/j.immuni.2024.03.020] [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: 04/05/2023] [Revised: 02/13/2024] [Accepted: 03/26/2024] [Indexed: 04/20/2024]
Abstract
Signaling through Notch receptors intrinsically regulates tumor cell development and growth. Here, we studied the role of the Notch ligand Jagged2 on immune evasion in non-small cell lung cancer (NSCLC). Higher expression of JAG2 in NSCLC negatively correlated with survival. In NSCLC pre-clinical models, deletion of Jag2, but not Jag1, in cancer cells attenuated tumor growth and activated protective anti-tumor T cell responses. Jag2-/- lung tumors exhibited higher frequencies of macrophages that expressed immunostimulatory mediators and triggered T cell-dependent anti-tumor immunity. Mechanistically, Jag2 ablation promoted Nr4a-mediated induction of Notch ligands DLL1/4 on cancer cells. DLL1/4-initiated Notch1/2 signaling in macrophages induced the expression of transcription factor IRF4 and macrophage immunostimulatory functionality. IRF4 expression was required for the anti-tumor effects of Jag2 deletion in lung tumors. Antibody targeting of Jagged2 inhibited tumor growth and activated IRF4-driven macrophage-mediated anti-tumor immunity. Thus, Jagged2 orchestrates immunosuppressive systems in NSCLC that can be overcome to incite macrophage-mediated anti-tumor immunity.
Collapse
Affiliation(s)
- Jay K Mandula
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Rachel V Jimenez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Darwin Chang
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Eslam Mohamed
- California Northstate University, Elk Grove, CA 95757, USA
| | - Shiun Chang
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Yu Cao
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Carmen M Anadon
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27708, USA
| | - Sae Bom Lee
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Satyajit Das
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Léo Rocha-Munguba
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Vincent M Pham
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Roger Li
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Ahmad A Tarhini
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA; Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Muhammad Furqan
- Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | | | | | - Carlos M Moran-Segura
- Advanced Analytical and Digital Laboratory, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jonathan Nguyen
- Advanced Analytical and Digital Laboratory, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Bradford Perez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Douglas J Kojetin
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alyssa Obermayer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Ann Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Timothy I Shaw
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA; Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jose R Conejo-Garcia
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27708, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
8
|
Oliveira RHDM, Annex BH, Popel AS. Endothelial cells signaling and patterning under hypoxia: a mechanistic integrative computational model including the Notch-Dll4 pathway. Front Physiol 2024; 15:1351753. [PMID: 38455844 PMCID: PMC10917925 DOI: 10.3389/fphys.2024.1351753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Introduction: Several signaling pathways are activated during hypoxia to promote angiogenesis, leading to endothelial cell patterning, interaction, and downstream signaling. Understanding the mechanistic signaling differences between endothelial cells under normoxia and hypoxia and their response to different stimuli can guide therapies to modulate angiogenesis. We present a novel mechanistic model of interacting endothelial cells, including the main pathways involved in angiogenesis. Methods: We calibrate and fit the model parameters based on well-established modeling techniques that include structural and practical parameter identifiability, uncertainty quantification, and global sensitivity. Results: Our results indicate that the main pathways involved in patterning tip and stalk endothelial cells under hypoxia differ, and the time under hypoxia interferes with how different stimuli affect patterning. Additionally, our simulations indicate that Notch signaling might regulate vascular permeability and establish different Nitric Oxide release patterns for tip/stalk cells. Following simulations with various stimuli, our model suggests that factors such as time under hypoxia and oxygen availability must be considered for EC pattern control. Discussion: This project provides insights into the signaling and patterning of endothelial cells under various oxygen levels and stimulation by VEGFA and is our first integrative approach toward achieving EC control as a method for improving angiogenesis. Overall, our model provides a computational framework that can be built on to test angiogenesis-related therapies by modulation of different pathways, such as the Notch pathway.
Collapse
Affiliation(s)
| | - Brian H. Annex
- Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Aleksander S. Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
9
|
Sunshine HL, Cicchetto AC, Kaczor-Urbanowicz KE, Ma F, Pi D, Symons C, Turner M, Shukla V, Christofk HR, Vallim TA, Iruela-Arispe ML. Endothelial Jagged1 levels and distribution are post-transcriptionally controlled by ZFP36 decay proteins. Cell Rep 2024; 43:113627. [PMID: 38157296 PMCID: PMC10884959 DOI: 10.1016/j.celrep.2023.113627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/02/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
Vascular morphogenesis requires a delicate gradient of Notch signaling controlled, in part, by the distribution of ligands (Dll4 and Jagged1). How Jagged1 (JAG1) expression is compartmentalized in the vascular plexus remains unclear. Here, we show that Jag1 mRNA is a direct target of zinc-finger protein 36 (ZFP36), an RNA-binding protein involved in mRNA decay that we find robustly induced by vascular endothelial growth factor (VEGF). Endothelial cells lacking ZFP36 display high levels of JAG1 and increase angiogenic sprouting in vitro. Furthermore, mice lacking Zfp36 in endothelial cells display mispatterned and increased levels of JAG1 in the developing retinal vascular plexus. Abnormal levels of JAG1 at the sprouting front alters NOTCH1 signaling, increasing the number of tip cells, a phenotype that is rescued by imposing haploinsufficiency of Jag1. Our findings reveal an important feedforward loop whereby VEGF stimulates ZFP36, consequently suppressing Jag1 to enable adequate levels of Notch signaling during sprouting angiogenesis.
Collapse
Affiliation(s)
- Hannah L Sunshine
- Molecular, Cellular, and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew C Cicchetto
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Karolina Elżbieta Kaczor-Urbanowicz
- Center for Oral and Head/Neck Oncology Research, UCLA Biosystems & Function, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095-1668, USA; UCLA Section of Orthodontics, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Feiyang Ma
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Danielle Pi
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Chloe Symons
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Martin Turner
- Immunology Programme, The Babraham Institute, CB22 3AT Cambridge, UK
| | - Vipul Shukla
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Center for Human Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Heather R Christofk
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095-1606, USA
| | - Thomas A Vallim
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095-1606, USA
| | - M Luisa Iruela-Arispe
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
10
|
Guo Y, Zhang S, Wang D, Heng BC, Deng X. Role of cell rearrangement and related signaling pathways in the dynamic process of tip cell selection. Cell Commun Signal 2024; 22:24. [PMID: 38195565 PMCID: PMC10777628 DOI: 10.1186/s12964-023-01364-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/25/2023] [Indexed: 01/11/2024] Open
Abstract
Angiogenesis is a complex, highly-coordinated and multi-step process of new blood vessel formation from pre-existing blood vessels. When initiated, the sprouting process is spearheaded by the specialized endothelial cells (ECs) known as tip cells, which guide the organization of accompanying stalk cells and determine the function and morphology of the finally-formed blood vessels. Recent studies indicate that the orchestration and coordination of angiogenesis involve dynamic tip cell selection, which is the competitive selection of cells to lead the angiogenic sprouts. Therefore, this review attempt to summarize the underlying mechanisms involved in tip cell specification in a dynamic manner to enable readers to gain a systemic and overall understanding of tip cell formation, involving cooperative interaction of cell rearrangement with Notch and YAP/TAZ signaling. Various mechanical and chemical signaling cues are integrated to ensure the right number of cells at the right place during angiogenesis, thereby precisely orchestrating morphogenic functions that ensure correct patterning of blood vessels. Video Abstract.
Collapse
Affiliation(s)
- Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Shihan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Dandan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China.
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
- Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| |
Collapse
|
11
|
Dong Y, Ma G, Hou X, Han Y, Ding Z, Tang W, Chen L, Chen Y, Zhou B, Rao F, Lv K, Du C, Cao H. Kindlin-2 controls angiogenesis through modulating Notch1 signaling. Cell Mol Life Sci 2023; 80:223. [PMID: 37480504 PMCID: PMC11072286 DOI: 10.1007/s00018-023-04866-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/02/2023] [Accepted: 07/08/2023] [Indexed: 07/24/2023]
Abstract
Kindlin-2 is critical for development and homeostasis of key organs, including skeleton, liver, islet, etc., yet its role in modulating angiogenesis is unknown. Here, we report that sufficient KINDLIN-2 is extremely important for NOTCH-mediated physiological angiogenesis. The expression of KINDLIN-2 in HUVECs is significantly modulated by angiogenic factors such as vascular endothelial growth factor A or tumor necrosis factor α. A strong co-localization of CD31 and Kindlin-2 in tissue sections is demonstrated by immunofluorescence staining. Endothelial-cell-specific Kindlin-2 deletion embryos die on E10.5 due to hemorrhage caused by the impaired physiological angiogenesis. Experiments in vitro show that vascular endothelial growth factor A-induced multiple functions of endothelial cells, including migration, matrix proteolysis, morphogenesis and sprouting, are all strengthened by KINDLIN-2 overexpression and severely impaired in the absence of KINDLIN-2. Mechanistically, we demonstrate that KINDLIN-2 inhibits the release of Notch intracellular domain through binding to and maintaining the integrity of NOTCH1. The impaired angiogenesis and avascular retinas caused by KINDLIN-2 deficiency can be rescued by DAPT, an inhibitor of γ-secretase which releases the intracellular domain from NOTCH1. Moreover, we demonstrate that high glucose stimulated hyperactive angiogenesis by increasing KINDLIN-2 expression could be prevented by KINDLIN-2 knockdown, indicating Kindlin-2 as a potential therapeutic target in treatment of diabetic retinopathy. Our study for the first time demonstrates the significance of Kindlin-2 in determining Notch-mediated angiogenesis during development and highlights Kindlin-2 as the potential therapeutic target in angiogenic diseases, such as diabetic retinopathy.
Collapse
Affiliation(s)
- Yuechao Dong
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guixing Ma
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xiaoting Hou
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yingying Han
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhen Ding
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wanze Tang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Litong Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yangshan Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Bo Zhou
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Feng Rao
- Southern University of Science and Technology, Shenzhen, 518055, China
| | - Kaosheng Lv
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Changzheng Du
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
12
|
Gjølberg TT, Wik JA, Johannessen H, Krüger S, Bassi N, Christopoulos PF, Bern M, Foss S, Petrovski G, Moe MC, Haraldsen G, Fosse JH, Skålhegg BS, Andersen JT, Sundlisæter E. Antibody blockade of Jagged1 attenuates choroidal neovascularization. Nat Commun 2023; 14:3109. [PMID: 37253747 PMCID: PMC10229650 DOI: 10.1038/s41467-023-38563-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/08/2023] [Indexed: 06/01/2023] Open
Abstract
Antibody-based blocking of vascular endothelial growth factor (VEGF) reduces choroidal neovascularization (CNV) and retinal edema, rescuing vision in patients with neovascular age-related macular degeneration (nAMD). However, poor response and resistance to anti-VEGF treatment occurs. We report that targeting the Notch ligand Jagged1 by a monoclonal antibody reduces neovascular lesion size, number of activated phagocytes and inflammatory markers and vascular leakage in an experimental CNV mouse model. Additionally, we demonstrate that Jagged1 is expressed in mouse and human eyes, and that Jagged1 expression is independent of VEGF signaling in human endothelial cells. When anti-Jagged1 was combined with anti-VEGF in mice, the decrease in lesion size exceeded that of either antibody alone. The therapeutic effect was solely dependent on blocking, as engineering antibodies to abolish effector functions did not impair the therapeutic effect. Targeting of Jagged1 alone or in combination with anti-VEGF may thus be an attractive strategy to attenuate CNV-bearing diseases.
Collapse
Affiliation(s)
- Torleif Tollefsrud Gjølberg
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway
- Center of Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, 0450, Oslo, Norway
| | - Jonas Aakre Wik
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Department of Nutrition, Division of Molecular Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
| | - Hanna Johannessen
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Department of Pediatric Surgery, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Stig Krüger
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Nicola Bassi
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | | | - Malin Bern
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway
| | - Stian Foss
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway
| | - Goran Petrovski
- Center of Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, 0450, Oslo, Norway
| | - Morten C Moe
- Center of Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, 0450, Oslo, Norway
| | - Guttorm Haraldsen
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Johanna Hol Fosse
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Bjørn Steen Skålhegg
- Department of Nutrition, Division of Molecular Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway.
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway.
| | - Eirik Sundlisæter
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway.
| |
Collapse
|
13
|
Oliveira RHM, Annex BH, Popel AS. Endothelial cells signaling and patterning under hypoxia: a mechanistic integrative computational model including the Notch-Dll4 pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539270. [PMID: 37205581 PMCID: PMC10187169 DOI: 10.1101/2023.05.03.539270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Several signaling pathways are activated during hypoxia to promote angiogenesis, leading to endothelial cell patterning, interaction, and downstream signaling. Understanding the mechanistic signaling differences between normoxia and hypoxia can guide therapies to modulate angiogenesis. We present a novel mechanistic model of interacting endothelial cells, including the main pathways involved in angiogenesis. We calibrate and fit the model parameters based on well-established modeling techniques. Our results indicate that the main pathways involved in the patterning of tip and stalk endothelial cells under hypoxia differ, and the time under hypoxia affects how a reaction affects patterning. Interestingly, the interaction of receptors with Neuropilin1 is also relevant for cell patterning. Our simulations under different oxygen concentrations indicate time- and oxygen-availability-dependent responses for the two cells. Following simulations with various stimuli, our model suggests that factors such as period under hypoxia and oxygen availability must be considered for pattern control. This project provides insights into the signaling and patterning of endothelial cells under hypoxia, contributing to studies in the field.
Collapse
Affiliation(s)
- Rebeca Hannah M Oliveira
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| | - Brian H Annex
- Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| |
Collapse
|
14
|
Halawi A, El Kurdi AB, Vernon KA, Solhjou Z, Choi JY, Saad AJ, Younis NK, Elfekih R, Mohammed MT, Deban CA, Weins A, Abdi R, Riella LV, De Serres SA, Cravedi P, Greka A, Khoueiry P, Azzi JR. Uncovering a novel role of focal adhesion and interferon-gamma in cellular rejection of kidney allografts at single cell resolution. Front Immunol 2023; 14:1139358. [PMID: 37063857 PMCID: PMC10102512 DOI: 10.3389/fimmu.2023.1139358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/23/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundKidney transplant recipients are currently treated with nonspecific immunosuppressants that cause severe systemic side effects. Current immunosuppressants were developed based on their effect on T-cell activation rather than the underlying mechanisms driving alloimmune responses. Thus, understanding the role of the intragraft microenvironment will help us identify more directed therapies with lower side effects.MethodsTo understand the role of the alloimmune response and the intragraft microenvironment in cellular rejection progression, we conducted a Single nucleus RNA sequencing (snRNA-seq) on one human non-rejecting kidney allograft sample, one borderline sample, and T-cell mediated rejection (TCMR) sample (Banff IIa). We studied the differential gene expression and enriched pathways in different conditions, in addition to ligand-receptor (L-R) interactions.ResultsPathway analysis of T-cells in borderline sample showed enrichment for allograft rejection pathway, suggesting that the borderline sample reflects an early rejection. Hence, this allows for studying the early stages of cellular rejection. Moreover, we showed that focal adhesion (FA), IFNg pathways, and endomucin (EMCN) were significantly upregulated in endothelial cell clusters (ECs) of borderline compared to ECs TCMR. Furthermore, we found that pericytes in TCMR seem to favor endothelial permeability compared to borderline. Similarly, T-cells interaction with ECs in borderline differs from TCMR by involving DAMPS-TLRs interactions.ConclusionOur data revealed novel roles of T-cells, ECs, and pericytes in cellular rejection progression, providing new clues on the pathophysiology of allograft rejection.
Collapse
Affiliation(s)
- Ahmad Halawi
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Abdullah B. El Kurdi
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Zhabiz Solhjou
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Scripps Clinic Medical Group, San Diego, CA, United States
| | - John Y. Choi
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Anis J. Saad
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Nour K. Younis
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Rania Elfekih
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Mostafa Tawfeek Mohammed
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Clinical Pathology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Christa A. Deban
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Astrid Weins
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Reza Abdi
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Leonardo V. Riella
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
| | - Sasha A. De Serres
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Paolo Cravedi
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anna Greka
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Pierre Khoueiry
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jamil R. Azzi
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Jamil R. Azzi,
| |
Collapse
|
15
|
Hasan SS, Fischer A. Notch Signaling in the Vasculature: Angiogenesis and Angiocrine Functions. Cold Spring Harb Perspect Med 2023; 13:cshperspect.a041166. [PMID: 35667708 PMCID: PMC9899647 DOI: 10.1101/cshperspect.a041166] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Formation of a functional blood vessel network is a complex process tightly controlled by pro- and antiangiogenic signals released within the local microenvironment or delivered through the bloodstream. Endothelial cells precisely integrate such temporal and spatial changes in extracellular signals and generate an orchestrated response by modulating signaling transduction, gene expression, and metabolism. A key regulator in vessel formation is Notch signaling, which controls endothelial cell specification, proliferation, migration, adhesion, and arteriovenous differentiation. This review summarizes the molecular biology of endothelial Notch signaling and how it controls angiogenesis and maintenance of the established, quiescent vasculature. In addition, recent progress in the understanding of Notch signaling in endothelial cells for controlling organ homeostasis by transcriptional regulation of angiocrine factors and its relevance to disease will be discussed.
Collapse
Affiliation(s)
- Sana S Hasan
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Andreas Fischer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Institute for Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany.,European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
16
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
17
|
Wang X, Zhao J, Wang X, Zhang J, Wang Y, Wang X, Jia S, Shi N, Lu M, Su H, Zhang J, Jiang D. Bacterial cellulose membrane combined with BMSCs promotes wound healing by activating the notch signaling pathway. Front Surg 2023; 9:1027067. [PMID: 36726958 PMCID: PMC9885103 DOI: 10.3389/fsurg.2022.1027067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/19/2022] [Indexed: 01/19/2023] Open
Abstract
Objective The bacterial cellulose membrane (BCM) has been widely studied and applied as a new biomaterial for wound healing, but causes pain with frequent dressing changes. Local application of bone marrow mesenchymal stem cells (BMSCs) requires a niche. Furthermore, the effect and mechanism of the BCM combined with BMSCs have not been reported. Methods Morphological and chemical identifications of BCMs were investigated by porosity analyses, scanning electron microscopy, and Fourier-transform infrared spectroscopy. Biological wound dressings (BWDs) were prepared by the BCM in combination with BMSCs. The biological effects of BWDs on human dermal fibroblast (HDF) and VEGF-A in human vascular endothelial cells (HuVECs) were detected in vitro, and the effect of BWDs on acute wounds in mice was detected in vivo. Collagen and angiogenesis were evaluated through hematoxylin-eosin staining and Masson staining. The expressions of COL-1 and VEGF-A and the activation of the Notch signaling pathway in vivo and in vitro were detected by quantitative reverse-transcriptase polymerase chain reaction. Results The BCM had a nanoscale structure and provided a partial niche for the survival and proliferation of BMSCs. BWDs were successfully prepared and regulated the biological behaviors of wound healing-related cells in vitro and upregulated the expressions of COL-1 in HDF and VEGF-A in HuVECs. BWDs promoted wound healing by increasing collagen type I synthesis and angiogenesis in acute wounds in mice. Conclusions BWDs prepared by the combination of nanomaterial BCMs and BMSCs facilitated acute wound healing, which may be regulated by activating the Notch signaling pathway.
Collapse
Affiliation(s)
- Xiaoyang Wang
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Jie Zhao
- Emergency Medicine Center, The Second Hospital of Shandong University, Jinan, China
| | - Xiaochuan Wang
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Jingjuan Zhang
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Yi Wang
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Xinyue Wang
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Shanshan Jia
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Nian Shi
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Meiqi Lu
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Hongxia Su
- Shandong Nameide Biotechnology Limited Company, Jinan, China
| | - Jixun Zhang
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China,Correspondence: Jixun Zhang Duyin Jiang
| | - Duyin Jiang
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, China,Emergency Medicine Center, The Second Hospital of Shandong University, Jinan, China,Correspondence: Jixun Zhang Duyin Jiang
| |
Collapse
|
18
|
Seynhaeve ALB, Ten Hagen TLM. An In Vivo Model to Study Cell Migration in XYZ-T Dimension Followed by Whole-Mount Re-evaluation. Methods Mol Biol 2023; 2608:325-341. [PMID: 36653716 DOI: 10.1007/978-1-0716-2887-4_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Cell migration is a very dynamic process involving several chemical as well as biological interactions with other cells and the environment. Several models exist to study cell migration ranging from simple 2D in vitro cultures to more demanding 3D multicellular assays, to complex evaluation in animals. High-resolution 4D (XYZ, spatial + T, time dimension) intravital imaging using transgenic animals with a fluorescent label in cells of interest is a powerful tool to study cell migration in the correct environment. Here we describe an advanced dorsal skinfold chamber model to study endothelial cell and pericyte migration and association.
Collapse
Affiliation(s)
- Ann L B Seynhaeve
- Laboratory Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, the Netherlands.
| | - Timo L M Ten Hagen
- Laboratory Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
19
|
Glioma diagnosis and therapy: Current challenges and nanomaterial-based solutions. J Control Release 2022; 352:338-370. [PMID: 36206948 DOI: 10.1016/j.jconrel.2022.09.065] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022]
Abstract
Glioma is often referred to as one of the most dreadful central nervous system (CNS)-specific tumors with rapidly-proliferating cancerous glial cells, accounting for nearly half of the brain tumors at an annual incidence rate of 30-80 per a million population. Although glioma treatment remains a significant challenge for researchers and clinicians, the rapid development of nanomedicine provides tremendous opportunities for long-term glioma therapy. However, several obstacles impede the development of novel therapeutics, such as the very tight blood-brain barrier (BBB), undesirable hypoxia, and complex tumor microenvironment (TME). Several efforts have been dedicated to exploring various nanoformulations for improving BBB permeation and precise tumor ablation to address these challenges. Initially, this article briefly introduces glioma classification and various pathogenic factors. Further, currently available therapeutic approaches are illustrated in detail, including traditional chemotherapy, radiotherapy, and surgical practices. Then, different innovative treatment strategies, such as tumor-treating fields, gene therapy, immunotherapy, and phototherapy, are emphasized. In conclusion, we summarize the article with interesting perspectives, providing suggestions for future glioma diagnosis and therapy improvement.
Collapse
|
20
|
Khan I, Siddiqui MN, Jameel F, Qazi REM, Salim A, Aslam S, Zaidi MB. Potential of stem cell seeded three-dimensional scaffold for regeneration of full-thickness skin wounds. Interface Focus 2022; 12:20220017. [PMID: 35996740 PMCID: PMC9372646 DOI: 10.1098/rsfs.2022.0017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/14/2022] [Indexed: 12/11/2022] Open
Abstract
Hypoxic wounds are tough to heal and are associated with chronicity, causing major healthcare burden. Available treatment options offer only limited success for accelerated and scarless healing. Traditional skin substitutes are widely used to improve wound healing, however, they lack proper vascularization. Mesenchymal stem cells (MSCs) offer improved wound healing; however, their poor retention, survival and adherence at the wound site negatively affect their therapeutic potential. The aim of this study is to enhance skin regeneration in a rat model of full-thickness dermal wound by transplanting genetically modified MSCs seeded on a three-dimensional collagen scaffold. Rat bone marrow MSCs were efficiently incorporated in the acellular collagen scaffold. Skin tissues with transplanted subcutaneous scaffolds were histologically analysed, while angiogenesis was assessed both at gene and protein levels. Our findings demonstrated that three-dimensional collagen scaffolds play a potential role in the survival and adherence of stem cells at the wound site, while modification of MSCs with jagged one gene provides a conducive environment for wound regeneration with improved proliferation, reduced inflammation and enhanced vasculogenesis. The results of this study represent an advanced targeted approach having the potential to be translated in clinical settings for targeted personalized therapy.
Collapse
Affiliation(s)
- Irfan Khan
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Marium Naz Siddiqui
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Fatima Jameel
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Rida-e-Maria Qazi
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Asmat Salim
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Shazmeen Aslam
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Midhat Batool Zaidi
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
21
|
Jiang N, Hu Y, Wang M, Zhao Z, Li M. The Notch Signaling Pathway Contributes to Angiogenesis and Tumor Immunity in Breast Cancer. BREAST CANCER: TARGETS AND THERAPY 2022; 14:291-309. [PMID: 36193236 PMCID: PMC9526507 DOI: 10.2147/bctt.s376873] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/08/2022] [Indexed: 11/23/2022]
Abstract
Breast cancer in women is the first leading tumor in terms of incidence worldwide. Some subtypes of BC lack distinct molecular targets and exhibit therapeutic resistance; these patients have a poor prognosis. Thus, the search for new molecular targets is an ongoing challenge for BC therapy. The Notch signaling pathway is found in both vertebrates and invertebrates, and it is a highly conserved in the evolution of the species, controlling cellular fates such as death, proliferation, and differentiation. Numerous studies have shown that improper activation of Notch signaling may lead to excessive cell proliferation and cancer, with tumor-promoting and tumor-suppressive effects in various carcinomas. Thus, inhibitors of Notch signaling are actively being investigated for the treatment of various tumors. The role of Notch signaling in BC has been widely studied in recent years. There is a growing body of evidence suggesting that Notch signaling has a pro-oncogenic role in BC, and the tumor-promoting effect is largely a result of the diverse nature of tumor immunity. Immunological abnormality is also a factor involved in the pathogenesis of BC, suggesting that Notch signaling could be a target for BC immunotherapies. Furthermore, angiogenesis is essential for BC growth and metastasis, and the Notch signaling pathway has been implicated in angiogenesis, so studying the role of Notch signaling in BC angiogenesis will provide new prospects for the treatment of BC. We summarize the potential roles of the current Notch signaling pathway and its inhibitors in BC angiogenesis and the immune response in this review and describe the pharmacological targets of Notch signaling in BC, which may serve as a theoretical foundation for future research into exploring this pathway for novel BC therapies.
Collapse
Affiliation(s)
- Nina Jiang
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Ye Hu
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Meiling Wang
- Department of Breast Surgery, the Second Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Zuowei Zhao
- Department of Breast Surgery, the Second Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
- Correspondence: Zuowei Zhao, Department of Breast Surgery, the Second Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China, Tel +86-0411-84671291, Fax +86-0411-84671230, Email
| | - Man Li
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
- Man Li, Department of Oncology, the Second Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China, Tel +86-0411-84671291, Fax +86-0411-84671230, Email
| |
Collapse
|
22
|
Fan L, Liu H, Zhu G, Singh S, Yu Z, Wang S, Luo H, Liu S, Xu Y, Ge J, Jiang D, Pang J. Caspase-4/11 is critical for angiogenesis by repressing Notch1 signaling via inhibiting γ-secretase activity. Br J Pharmacol 2022; 179:4809-4828. [PMID: 35737588 DOI: 10.1111/bph.15904] [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: 11/16/2021] [Revised: 05/23/2022] [Accepted: 05/29/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Notch1 activation mediated by γ-secretase is critical for angiogenesis. GeneCards database predicted that Caspase-4 (CASP4, with murine ortholog CASP11) interacts with presenilin-1, the catalytic core of γ-secretase. Therefore, we investigated the role of CASP4/11 in angiogenesis. EXPERIMENTAL APPROACH In vivo, we studied the role of Casp11 in several angiogenesis mouse models using Casp11 wild-type and knockout mice. In vitro, we detected the effects of CASP4 on endothelial functions and Notch signaling by depleting or overexpressing CASP4 in human umbilical vein endothelial cells (HUVECs). The functional domain responsible for the binding of CASP4 and presenilin-1 was detected by mutagenesis and co-immunoprecipitation. KEY RESULTS Casp11 deficiency remarkably impaired adult angiogenesis in ischemic hindlimbs, melanoma xenografts and Matrigel plugs, but not the developmental angiogenesis of retina. Bone marrow transplantation revealed that the pro-angiogenic effect depended on CASP11 derived from non-hematopoietic cells. CASP4 expression was induced by inflammatory factors and CASP4 knockdown decreased cell viability, proliferation, migration and tube formation in HUVECs. Mechanistically, CASP4/11 deficiency increased Notch1 activation in vivo and in vitro, while CASP4 overexpression repressed Notch1 signaling in HUVECs. Moreover, CASP4 knockdown increased γ-secretase activity. γ-Secretase inhibitor DAPT restored the effects of CASP4 siRNA on Notch1 activation and angiogenesis in HUVECs. Notably, the catalytic activity of CASP4/11 was dispensable. Instead, CASP4 directly interacted with presenilin-1 through the caspase recruitment domain (CARD). CONCLUSIONS AND IMPLICATIONS These findings reveal a critical role of CASP4/11 in adult angiogenesis and make this molecule a promising therapeutic target for angiogenesis-related diseases in the future.
Collapse
Affiliation(s)
- Linlin Fan
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Hao Liu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guofu Zhu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shekhar Singh
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ze Yu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shumin Wang
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Hong Luo
- Department of Medical Laboratory, College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Shiying Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junbo Ge
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Dongyang Jiang
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinjiang Pang
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| |
Collapse
|
23
|
Niu H, Gao N, Dang Y, Guan Y, Guan J. Delivery of VEGF and delta-like 4 to synergistically regenerate capillaries and arterioles in ischemic limbs. Acta Biomater 2022; 143:295-309. [PMID: 35301145 PMCID: PMC9926495 DOI: 10.1016/j.actbio.2022.03.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/11/2022]
Abstract
Vascularization of the poorly vascularized limbs affected by critical limb ischemia (CLI) is necessary to salvage the limbs and avoid amputation. Effective vascularization requires forming not only capillaries, but also arterioles and vessel branching. These processes rely on the survival, migration and morphogenesis of endothelial cells in the ischemic limbs. Yet endothelial cell functions are impaired by the upregulated TGFβ. Herein, we developed an injectable hydrogel-based drug release system capable of delivering both VEGF and Dll4 to synergistically restore endothelial cellular functions, leading to accelerated formation of capillaries, arterioles and vessel branching. In vitro, the Dll4 and VEGF synergistically promoted the human arterial endothelial cell (HAEC) survival, migration, and formation of filopodial structure, lumens, and branches under the elevated TGFβ1 condition mimicking that of the ischemic limbs. The synergistic effect was resulted from activating VEGFR2, Notch-1 and Erk1/2 signaling pathways. After delivering the Dll4 and VEGF via an injectable and thermosensitive hydrogel to the ischemic mouse hindlimbs, 95% of blood perfusion was restored at day 14, significantly higher than delivery of Dll4 or VEGF only. The released Dll4 and VEGF significantly increased density of capillaries and arterioles, vessel branching point density, and proliferating cell density. Besides, the delivery of Dll4 and VEGF stimulated skeletal muscle regeneration and improved muscle function. Overall, the developed hydrogel-based Dll4 and VEGF delivery system promoted ischemic limb vascularization and muscle regeneration. STATEMENT OF SIGNIFICANCE: Effective vascularization of the poorly vascularized limbs affected by critical limb ischemia (CLI) requires forming not only capillaries, but also arterioles and vessel branching. These processes rely on the survival, migration and morphogenesis of endothelial cells. Yet endothelial cell functions are impaired by the upregulated TGFβ in the ischemic limbs. Herein, we developed an injectable hydrogel-based drug release system capable of delivering both VEGF and Dll4 to synergistically restore endothelial cell functions, leading to accelerated formation of capillaries, arterioles and vessel branching.
Collapse
Affiliation(s)
- Hong Niu
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis. St. Louis, MO, 63130, United States; Center of Regenerative Medicine, Washington University in St. Louis. St. Louis, MO, 63130, United States; Department of Materials Science and Engineering, Ohio State University. Columbus, OH, 43210, United States
| | - Ning Gao
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis. St. Louis, MO, 63130, United States; Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO, 63130, United States
| | - Yu Dang
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis. St. Louis, MO, 63130, United States; Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO, 63130, United States
| | - Ya Guan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis. St. Louis, MO, 63130, United States; Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO, 63130, United States
| | - Jianjun Guan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis. St. Louis, MO, 63130, United States; Center of Regenerative Medicine, Washington University in St. Louis. St. Louis, MO, 63130, United States; Department of Materials Science and Engineering, Ohio State University. Columbus, OH, 43210, United States; Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO, 63130, United States.
| |
Collapse
|
24
|
Tefft JB, Bays JL, Lammers A, Kim S, Eyckmans J, Chen CS. Notch1 and Notch3 coordinate for pericyte-induced stabilization of vasculature. Am J Physiol Cell Physiol 2022; 322:C185-C196. [PMID: 34878922 PMCID: PMC8791789 DOI: 10.1152/ajpcell.00320.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The Notch pathway regulates complex patterning events in many species and is critical for the proper formation and function of the vasculature. Despite this importance, how the various components of the Notch pathway work in concert is still not well understood. For example, NOTCH1 stabilizes homotypic endothelial junctions, but the role of NOTCH1 in heterotypic interactions is not entirely clear. NOTCH3, on the other hand, is essential for heterotypic interactions of pericytes with the endothelium, but how NOTCH3 signaling in pericytes impacts the endothelium remains elusive. Here, we use in vitro vascular models to investigate whether pericyte-induced stabilization of the vasculature requires the cooperation of NOTCH1 and NOTCH3. We observe that both pericyte NOTCH3 and endothelial NOTCH1 are required for the stabilization of the endothelium. Loss of either NOTCH3 or NOTCH1 decreases the accumulation of VE-cadherin at endothelial adherens junctions and increases the frequency of wider, more motile junctions. We found that DLL4 was the key ligand for simulating NOTCH1 activation in endothelial cells and observed that DLL4 expression in pericytes is dependent on NOTCH3. Altogether, these data suggest that an interplay between pericyte NOTCH3 and endothelial NOTCH1 is critical for pericyte-induced vascular stabilization.
Collapse
Affiliation(s)
- Juliann B. Tefft
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Jennifer L. Bays
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Alex Lammers
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Sudong Kim
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Jeroen Eyckmans
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Christopher S. Chen
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| |
Collapse
|
25
|
Swaminathan B, Youn SW, Naiche LA, Du J, Villa SR, Metz JB, Feng H, Zhang C, Kopan R, Sims PA, Kitajewski JK. Endothelial Notch signaling directly regulates the small GTPase RND1 to facilitate Notch suppression of endothelial migration. Sci Rep 2022; 12:1655. [PMID: 35102202 PMCID: PMC8804000 DOI: 10.1038/s41598-022-05666-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/07/2022] [Indexed: 11/24/2022] Open
Abstract
To control sprouting angiogenesis, endothelial Notch signaling suppresses tip cell formation, migration, and proliferation while promoting barrier formation. Each of these responses may be regulated by distinct Notch-regulated effectors. Notch activity is highly dynamic in sprouting endothelial cells, while constitutive Notch signaling drives homeostatic endothelial polarization, indicating the need for both rapid and constitutive Notch targets. In contrast to previous screens that focus on genes regulated by constitutively active Notch, we characterized the dynamic response to Notch. We examined transcriptional changes from 1.5 to 6 h after Notch signal activation via ligand-specific or EGTA induction in cultured primary human endothelial cells and neonatal mouse brain. In each combination of endothelial type and Notch manipulation, transcriptomic analysis identified distinct but overlapping sets of rapidly regulated genes and revealed many novel Notch target genes. Among the novel Notch-regulated signaling pathways identified were effectors in GPCR signaling, notably, the constitutively active GTPase RND1. In endothelial cells, RND1 was shown to be a novel direct Notch transcriptional target and required for Notch control of sprouting angiogenesis, endothelial migration, and Ras activity. We conclude that RND1 is directly regulated by endothelial Notch signaling in a rapid fashion in order to suppress endothelial migration.
Collapse
Affiliation(s)
- Bhairavi Swaminathan
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Seock-Won Youn
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - L A Naiche
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Jing Du
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Stephanie R Villa
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Jordan B Metz
- Department of Systems Biology, Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
| | - Huijuan Feng
- Department of Systems Biology, Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
| | - Chaolin Zhang
- Department of Systems Biology, Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
| | - Raphael Kopan
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Peter A Sims
- Department of Systems Biology, Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
| | - Jan K Kitajewski
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
26
|
Chesnais F, Hue J, Roy E, Branco M, Stokes R, Pellon A, Le Caillec J, Elbahtety E, Battilocchi M, Danovi D, Veschini L. High content Image Analysis to study phenotypic heterogeneity in endothelial cell monolayers. J Cell Sci 2022; 135:273879. [PMID: 34982151 DOI: 10.1242/jcs.259104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/15/2021] [Indexed: 11/20/2022] Open
Abstract
Endothelial cells (EC) are heterogeneous across and within tissues, reflecting distinct, specialised functions. EC heterogeneity has been proposed to underpin EC plasticity independently from vessel microenvironments. However, heterogeneity driven by contact-dependent or short-range cell-cell crosstalk cannot be evaluated with single cell transcriptomic approaches as spatial and contextual information is lost. Nonetheless, quantification of EC heterogeneity and understanding of its molecular drivers is key to developing novel therapeutics for cancer, cardiovascular diseases and for revascularisation in regenerative medicine. Here, we developed an EC profiling tool (ECPT) to examine individual cells within intact monolayers. We used ECPT to characterise different phenotypes in arterial, venous and microvascular EC populations. In line with other studies, we measured heterogeneity in terms of cell cycle, proliferation, and junction organisation. ECPT uncovered a previously under-appreciated single-cell heterogeneity in NOTCH activation. We correlated cell proliferation with different NOTCH activation states at the single cell and population levels. The positional and relational information extracted with our novel approach is key to elucidating the molecular mechanisms underpinning EC heterogeneity.
Collapse
Affiliation(s)
- Francois Chesnais
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Jonas Hue
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Errin Roy
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK
| | - Marco Branco
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Ruby Stokes
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Aize Pellon
- Centre for host-microbiome interactions, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Juliette Le Caillec
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Eyad Elbahtety
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Matteo Battilocchi
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK
| | - Davide Danovi
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK.,bit.bio, Babraham Research Campus, The Dorothy Hodgkin Building, Cambridge CB22 3FH, UK
| | - Lorenzo Veschini
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
27
|
Ontoria-Oviedo I, Földes G, Tejedor S, Panadero J, Kitani T, Vázquez A, Wu JC, Harding SE, Sepúlveda P. Modeling Transposition of the Great Arteries with Patient-Specific Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms222413270. [PMID: 34948064 PMCID: PMC8705900 DOI: 10.3390/ijms222413270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
The dextro-transposition of the great arteries (d-TGA) is one of the most common congenital heart diseases. To identify biological processes that could be related to the development of d-TGA, we established induced pluripotent stem cell (iPSC) lines from two patients with d-TGA and from two healthy subjects (as controls) and differentiated them into endothelial cells (iPSC-ECs). iPSC-EC transcriptome profiling and bioinformatics analysis revealed differences in the expression level of genes involved in circulatory system and animal organ development. iPSC-ECs from patients with d-TGA showed impaired ability to develop tubular structures in an in vitro capillary-like tube formation assay, and interactome studies revealed downregulation of biological processes related to Notch signaling, circulatory system development and angiogenesis, pointing to alterations in vascular structure development. Our study provides an iPSC-based cellular model to investigate the etiology of d-TGA.
Collapse
Affiliation(s)
- Imelda Ontoria-Oviedo
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (S.T.); (A.V.)
- Correspondence: (I.O.-O.); (P.S.); Tel.: +34-96-1246632 (I.O.-O.); +34-96-1246635 (P.S.)
| | - Gabor Földes
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (G.F.); (S.E.H.)
- Heart and Vascular Center, Semmelweis University, H1122 Budapest, Hungary
| | - Sandra Tejedor
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (S.T.); (A.V.)
| | - Joaquín Panadero
- IGENOMIX S.L., Edificios Europark, Parque Tecnológico, 46980 Paterna, Spain;
| | - Tomoya Kitani
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.K.); (J.C.W.)
| | - Alejandro Vázquez
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (S.T.); (A.V.)
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.K.); (J.C.W.)
| | - Sian E. Harding
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (G.F.); (S.E.H.)
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (S.T.); (A.V.)
- Correspondence: (I.O.-O.); (P.S.); Tel.: +34-96-1246632 (I.O.-O.); +34-96-1246635 (P.S.)
| |
Collapse
|
28
|
Muley A, Kim Uh M, Salazar-De Simone G, Swaminathan B, James JM, Murtomaki A, Youn SW, McCarron JD, Kitajewski C, Gnarra Buethe M, Riitano G, Mukouyama YS, Kitajewski J, Shawber CJ. Unique functions for Notch4 in murine embryonic lymphangiogenesis. Angiogenesis 2021; 25:205-224. [PMID: 34665379 PMCID: PMC9054879 DOI: 10.1007/s10456-021-09822-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 10/08/2021] [Indexed: 11/08/2022]
Abstract
In mice, embryonic dermal lymphatic development is well understood and used to study gene functions in lymphangiogenesis. Notch signaling is an evolutionarily conserved pathway that modulates cell fate decisions, which has been shown to both inhibit and promote dermal lymphangiogenesis. Here, we demonstrate distinct roles for Notch4 signaling versus canonical Notch signaling in embryonic dermal lymphangiogenesis. Actively growing embryonic dermal lymphatics expressed NOTCH1, NOTCH4, and DLL4 which correlated with Notch activity. In lymphatic endothelial cells (LECs), DLL4 activation of Notch induced a subset of Notch effectors and lymphatic genes, which were distinctly regulated by Notch1 and Notch4 activation. Treatment of LECs with VEGF-A or VEGF-C upregulated Dll4 transcripts and differentially and temporally regulated the expression of Notch1 and Hes/Hey genes. Mice nullizygous for Notch4 had an increase in the closure of the lymphangiogenic fronts which correlated with reduced vessel caliber in the maturing lymphatic plexus at E14.5 and reduced branching at E16.5. Activation of Notch4 suppressed LEC migration in a wounding assay significantly more than Notch1, suggesting a dominant role for Notch4 in regulating LEC migration. Unlike Notch4 nulls, inhibition of canonical Notch signaling by expressing a dominant negative form of MAML1 (DNMAML) in Prox1+ LECs led to increased lymphatic density consistent with an increase in LEC proliferation, described for the loss of LEC Notch1. Moreover, loss of Notch4 did not affect LEC canonical Notch signaling. Thus, we propose that Notch4 signaling and canonical Notch signaling have distinct functions in the coordination of embryonic dermal lymphangiogenesis.
Collapse
Affiliation(s)
- Ajit Muley
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Minji Kim Uh
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA.,Department of Pharmacology, Columbia University Medical Center, New York, NY, 10032, USA
| | | | - Bhairavi Swaminathan
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Jennifer M James
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Aino Murtomaki
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA.,Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu, 8, 00290, Helsinki, Finland.,Translational Cancer Medicine Program, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Seock Won Youn
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Joseph D McCarron
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Chris Kitajewski
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Maria Gnarra Buethe
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Gloria Riitano
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA.,Departments of Molecular Medicine and Experimental Medicine, Sapienza University, 00185, Rome, Italy
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Carrie J Shawber
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA. .,Department of Surgery, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
29
|
Xiu M, Zeng X, Shan R, Wen W, Li J, Wan R. Targeting Notch4 in Cancer: Molecular Mechanisms and Therapeutic Perspectives. Cancer Manag Res 2021; 13:7033-7045. [PMID: 34526819 PMCID: PMC8436177 DOI: 10.2147/cmar.s315511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/03/2021] [Indexed: 12/28/2022] Open
Abstract
The dysregulation of Notch signaling is found in many cancers and is closely related to cancer progression. As an important Notch receptor, abnormal Notch4 expression affects several tumor-cell behaviors, including stemness, the epithelial-mesenchymal transition, radio/chemoresistance and angiogenesis. In order to inhibit the oncogenic effects of Notch4 activation, several methods for targeting Notch4 signaling have been proposed. In this review, we summarize the known molecular mechanisms through which Notch4 affects cancer progression. Finally, we discuss potential Notch4-targeting therapeutic strategies as a reference for future research.
Collapse
Affiliation(s)
- Mengxi Xiu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China.,Second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Xiaohong Zeng
- Imaging Department, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| | - Renfeng Shan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| | - Wu Wen
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| | - Jianfeng Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| | - Renhua Wan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
30
|
Abdul Kareem N, Aijaz A, Jeschke MG. Stem Cell Therapy for Burns: Story so Far. Biologics 2021; 15:379-397. [PMID: 34511880 PMCID: PMC8418374 DOI: 10.2147/btt.s259124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
Burn injuries affect approximately 11 million people annually, with fatalities amounting up to 180,000. Burn injuries constitute a global health issue associated with high morbidity and mortality. Recent years have seen advancements in regenerative medicine for burn wound healing encompassing stem cells and stem cell-derived products such as exosomes and conditioned media with promising results compared to current treatment approaches. Sources of stem cells used for treatment vary ranging from hair follicle stem cells, embryonic stem cells, umbilical cord stem cells, to mesenchymal stem cells, such as adipose-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, and even stem cells harvested from discarded burn tissue. Stem cells utilize various pathways for wound healing, such as PI3/AKT pathway, WNT-β catenin pathway, TGF-β pathway, Notch and Hedgehog signaling pathway. Due to the paracrine signaling mechanism of stem cells, exosomes and conditioned media derived from stem cells have also been utilized in burn wound therapy. As exosomes and conditioned media are cell-free therapy and contain various biomolecules that facilitate wound healing, they are gaining popularity as an alternative treatment strategy with significant improvement in outcomes. The treatment is provided either as direct injections or embedded in a natural/artificial scaffold. This paper reviews in detail the different sources of stem cells, stem cell-derived products, their efficacy in burn wound repair, associated signaling pathways and modes of delivery for wound healing.
Collapse
Affiliation(s)
| | - Ayesha Aijaz
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Marc G Jeschke
- Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada.,Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| |
Collapse
|
31
|
Immobilization of Jagged1 Enhances Vascular Smooth Muscle Cells Maturation by Activating the Notch Pathway. Cells 2021; 10:cells10082089. [PMID: 34440858 PMCID: PMC8391929 DOI: 10.3390/cells10082089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
In Notch signaling, the Jagged1-Notch3 ligand-receptor pairing is implicated for regulating the phenotype maturity of vascular smooth muscle cells. However, less is known about the role of Jagged1 presentation strategy in this regulation. In this study, we used bead-immobilized Jagged1 to direct phenotype control of primary human coronary artery smooth muscle cells (HCASMC), and to differentiate embryonic multipotent mesenchymal progenitor (10T1/2) cell towards a vascular lineage. This Jagged1 presentation strategy was sufficient to activate the Notch transcription factor HES1 and induce early-stage contractile markers, including smooth muscle α-actin and calponin in HCASMCs. Bead-bound Jagged1 was unable to regulate the late-stage markers myosin heavy chain and smoothelin; however, serum starvation and TGFβ1 were used to achieve a fully contractile smooth muscle cell. When progenitor 10T1/2 cells were used for Notch3 signaling, pre-differentiation with TGFβ1 was required for a robust Jagged1 specific response, suggesting a SMC lineage commitment was necessary to direct SMC differentiation and maturity. The presence of a magnetic tension force to the ligand-receptor complex was evaluated for signaling efficacy. Magnetic pulling forces downregulated HES1 and smooth muscle α-actin in both HCASMCs and progenitor 10T1/2 cells. Taken together, this study demonstrated that (i) bead-bound Jagged1 was sufficient to activate Notch3 and promote SMC differentiation/maturation and (ii) magnetic pulling forces did not activate Notch3, suggesting the bead alone was able to provide necessary clustering or traction forces for Notch activation. Notch is highly context-dependent; therefore, these findings provide insights to improve biomaterial-driven Jagged1 control of SMC behavior.
Collapse
|
32
|
Zhu Y, Hu J, Du X, Fang Q, Zhou Y, Chen K. Correlation of serum delta-like ligand-4 level with the severity of diabetic retinopathy. BMC Endocr Disord 2021; 21:157. [PMID: 34362349 PMCID: PMC8344193 DOI: 10.1186/s12902-021-00814-6] [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] [Received: 03/22/2021] [Accepted: 07/21/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is one of the most serious microvascular complications of type 2 diabetes mellitus (T2DM). Delta-like ligand-4 (DLL4) maintains the normal physiological microenvironment of the retina. However, the relationship between the level of DLL4 and the severity of DR remains unclear. METHODS We retrospectively analyzed serum DLL4 levels and other laboratory and clinical data in 94 T2DM patients (35 patients without DR [NDR], 32 with non-proliferative DR [NPDR], 27 with proliferative DR [PDR]), and 30 healthy controls. RESULTS The serum DLL4 level was significantly greater in the NDR group (43.38 ± 16.23 pg/mL), NPDR group (56.57 ± 25.89 pg/mL), and PDR group (74.97 ± 25.28 pg/mL) than in the healthy controls (29.9 ± 8.92 pg/mL; all p < 0.05). Among T2DM patients, the level of DLL4 increased as the severity of DR increased (p < 0.05). Logistic regression analysis demonstrated that DR was positively associated with DLL4, glycosylated hemoglobin (HbA1c), fasting blood glucose (FBG), and duration of T2DM (all p < 0.05). Consistently, receiver operating characteristic (ROC) curve analysis also indicated that DLL4 was a potential candidate biomarker for identifying the severity of DR. CONCLUSIONS T2DM patients, especially those with DR, have increased serum levels of DLL4. DLL4 may be used as a biomarker and an independent risk factor for DR, and targeting DLL4 may be a potential therapy in patients with DR.
Collapse
Affiliation(s)
- Yan Zhu
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, No.188 Shizi Road, Suzhou, China
| | - Jingcheng Hu
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, No.188 Shizi Road, Suzhou, China
| | - Xuan Du
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, No.188 Shizi Road, Suzhou, China
| | - Qionglei Fang
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, No.188 Shizi Road, Suzhou, China
| | - Yingyi Zhou
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, No.188 Shizi Road, Suzhou, China.
| | - Ke Chen
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, No.188 Shizi Road, Suzhou, China.
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, No.188 Shizi Road, Suzhou, China.
| |
Collapse
|
33
|
Brzozowa-Zasada M. The role of Notch ligand, Delta-like ligand 4 (DLL4), in cancer angiogenesis—implications for therapy. Eur Surg 2021. [DOI: 10.1007/s10353-021-00707-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Summary
Background
It is generally accepted that angiogenesis is a complex and tightly regulated process characterized by the growth of blood vessels from existing vasculature. Activation of the Notch signalling pathway affects multiple aspects of vascular development. One of the components of the Notch signalling pathway, Delta-like ligand 4 (DLL4), has recently appeared as a critical regulator of tumour angiogenesis and thus as a promising therapeutic target.
Methods
This review article includes available data from peer-reviewed publications associated with the role of DLL4 in cancer angiogenesis. Searches were performed in PubMed, EMBASE, Google Scholar and Web of Science using the terms “tumour angiogenesis”, “DLL4”, “Notch signalling” and “anti-cancer therapy”.
Results
The survival curves of cancer patients revealed that the patients with high DLL4 expression levels had significantly shorter survival times than the patients with low DLL4 expression. Moreover, a positive correlation was also identified between DLL4 and VEGF receptorsʼ expression levels. It seems that inhibition of DLL4 may exert potent growth inhibitory effects on some tumours resistant to anti-VEGF therapies. A great number of blocking agents of DLL4/Notch signalling including anti-DLL4 antibodies, DNA vaccination, Notch antibodies and gamma-secretase inhibitors have been studied in preclinical tumour models.
Conclusion
DLL4 seems to be a promising target in anti-cancer therapy. Nevertheless, the careful evaluation of adverse effects on normal physiological processes in relation to therapeutic doses of anti-DLL4 drugs will be significant for advancement of DLL4 blocking agents in clinical oncology.
Collapse
|
34
|
Starosta A, Konieczny P. Therapeutic aspects of cell signaling and communication in Duchenne muscular dystrophy. Cell Mol Life Sci 2021; 78:4867-4891. [PMID: 33825942 PMCID: PMC8233280 DOI: 10.1007/s00018-021-03821-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/26/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating chromosome X-linked disease that manifests predominantly in progressive skeletal muscle wasting and dysfunctions in the heart and diaphragm. Approximately 1/5000 boys and 1/50,000,000 girls suffer from DMD, and to date, the disease is incurable and leads to premature death. This phenotypic severity is due to mutations in the DMD gene, which result in the absence of functional dystrophin protein. Initially, dystrophin was thought to be a force transducer; however, it is now considered an essential component of the dystrophin-associated protein complex (DAPC), viewed as a multicomponent mechanical scaffold and a signal transduction hub. Modulating signal pathway activation or gene expression through epigenetic modifications has emerged at the forefront of therapeutic approaches as either an adjunct or stand-alone strategy. In this review, we propose a broader perspective by considering DMD to be a disease that affects myofibers and muscle stem (satellite) cells, as well as a disorder in which abrogated communication between different cell types occurs. We believe that by taking this systemic view, we can achieve safe and holistic treatments that can restore correct signal transmission and gene expression in diseased DMD tissues.
Collapse
Affiliation(s)
- Alicja Starosta
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland
| | - Patryk Konieczny
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| |
Collapse
|
35
|
Qiu TY, Huang J, Wang LP, Zhu BS. Inhibition of miR-200b Promotes Angiogenesis in Endothelial Cells by Activating The Notch Pathway. CELL JOURNAL 2021; 23:51-60. [PMID: 33650820 PMCID: PMC7944128 DOI: 10.22074/cellj.2021.7080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/05/2019] [Indexed: 12/27/2022]
Abstract
Objective Patients with diabetes mellitus frequently have chronic wounds or diabetic ulcers as a result of impaired
wound healing, which may lead to limb amputation. Human umbilical vein endothelial cell (HUVEC) dysfunction also
delays wound healing. Here, we investigated the mechanism of miR-200b in HUVECs under high glucose conditions
and the potential of miR-200b as a therapeutic target.
Materials and Methods In this experimental study, HUVECs were cultured with 5 or 30 mM glucose for 48 hours.
Cell proliferation was evaluated by CCK-8 assays. Cell mobility was tested by wound healing and Transwell assays.
Angiogenesis was analyzed in vitro Matrigel tube formation assays. Luciferase reporter assays were used to test the
binding of miR-200b with Notch1.
Results miR-200b expression was induced by high glucose treatment of HUVECs (P<0.01), and it significantly
repressed cell proliferation, migration, and tube formation (P<0.05). Notch1 was directly targeted and repressed by
miR-200b at both the mRNA and protein levels. Inhibition of miR-200b restored Notch1 expression (P<0.05) and
reactivated the Notch pathway. The effects of miR-200b inhibition in HUVECs could be reversed by treatment with a
Notch pathway inhibitor (P<0.05), indicating that the miR-200b/Notch axis modulates the proliferation, migration, and
tube formation ability of HUVECs.
Conclusion Inhibition of miR-200b activated the angiogenic ability of endothelial cells and promoted wound healing
through reactivation of the Notch pathway in vitro. miR-200b could be a promising therapeutic target for treating HUVEC
dysfunction.
Collapse
Affiliation(s)
- Tie-Ying Qiu
- Clinical Nursing Teaching and Research Section of the Second Xiangya Hospital, Changsha 410011, P.R. China
| | - Jin Huang
- Clinical Nursing Teaching and Research Section of the Second Xiangya Hospital, Changsha 410011, P.R. China
| | - Li-Ping Wang
- Clinical Nursing Teaching and Research Section of the Second Xiangya Hospital, Changsha 410011, P.R. China
| | - Bi-Song Zhu
- Organ Transplant Center, Xiangya Hospital, Central South University, Changsha 410008, P.R. China. Emails:
| |
Collapse
|
36
|
Insights into the mechanism of vascular endothelial cells on bone biology. Biosci Rep 2021; 41:227494. [PMID: 33403387 PMCID: PMC7816070 DOI: 10.1042/bsr20203258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/25/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022] Open
Abstract
In the skeletal system, blood vessels not only function as a conduit system for transporting gases, nutrients, metabolic waste, or cells but also provide multifunctional signal molecules regulating bone development, regeneration, and remodeling. Endothelial cells (ECs) in bone tissues, unlike in other organ tissues, are in direct contact with the pericytes of blood vessels, resulting in a closer connection with peripheral connective tissues. Close-contact ECs contribute to osteogenesis and osteoclastogenesis by secreting various cytokines in the paracrine or juxtacrine pathways. An increasing number of studies have revealed that extracellular vesicles (EVs) derived from ECs can directly regulate maturation process of osteoblasts and osteoclasts. The different pathways focus on targets at different distances, forming the basis of the intimate spatial and temporal link between bone tissue and blood vessels. Here, we provide a systematic review to elaborate on the function of ECs in bone biology and its underlying mechanisms based on three aspects: paracrine, EVs, and juxtacrine. This review proposes the possibility of a therapeutic strategy targeting blood vessels, as an adjuvant treatment for bone disorders.
Collapse
|
37
|
Reichrath J, Reichrath S. The Impact of Notch Signaling for Carcinogenesis and Progression of Nonmelanoma Skin Cancer: Lessons Learned from Cancer Stem Cells, Tumor Angiogenesis, and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1287:123-154. [PMID: 33034030 DOI: 10.1007/978-3-030-55031-8_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since many decades, nonmelanoma skin cancer (NMSCs) is the most common malignancy worldwide. Basal cell carcinomas (BCC) and squamous cell carcinomas (SCC) are the major types of NMSCs, representing approximately 70% and 25% of these neoplasias, respectively. Because of their continuously rising incidence rates, NMSCs represent a constantly increasing global challenge for healthcare, although they are in most cases nonlethal and curable (e.g., by surgery). While at present, carcinogenesis of NMSC is still not fully understood, the relevance of genetic and molecular alterations in several pathways, including evolutionary highly conserved Notch signaling, has now been shown convincingly. The Notch pathway, which was first developed during evolution in metazoans and that was first discovered in fruit flies (Drosophila melanogaster), governs cell fate decisions and many other fundamental processes that are of high relevance not only for embryonic development, but also for initiation, promotion, and progression of cancer. Choosing NMSC as a model, we give in this review a brief overview on the interaction of Notch signaling with important oncogenic and tumor suppressor pathways and on its role for several hallmarks of carcinogenesis and cancer progression, including the regulation of cancer stem cells, tumor angiogenesis, and senescence.
Collapse
Affiliation(s)
- Jörg Reichrath
- Department of Dermatology, Saarland University Medical Center, Homburg, Germany.
| | - Sandra Reichrath
- Department of Dermatology, Saarland University Medical Center, Homburg, Germany.,School of Health Professions, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
38
|
Alhayaza R, Haque E, Karbasiafshar C, Sellke FW, Abid MR. The Relationship Between Reactive Oxygen Species and Endothelial Cell Metabolism. Front Chem 2020; 8:592688. [PMID: 33330380 PMCID: PMC7732658 DOI: 10.3389/fchem.2020.592688] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) has been the leading cause of death for many decades, highlighting the importance of new research and treatments in the field. The role of hypoxia and subsequent free radical production [reactive oxygen species (ROS)] have become an area of particular interest in CVD. Interestingly, our laboratory and other laboratories have recently reported positive roles of subcellular ROS in modulating endothelial cell (EC) metabolism, proliferation, and angiogenesis. This bidirectional relationship between ROS and EC metabolism, as well as functional changes, continues to be an area of active research. Interestingly, ECs have been shown to rely on anaerobic processes for ATP generation, despite their direct access to oxygen. This paradox has proven to be beneficial as the major reliance on glycolysis produces ATP faster, preserves oxygen, and results in reduced ROS levels in contrast to oxidative phosphorylation. This review will address the relationship between ROS and carbohydrate, lipid, and nitrogen metabolism in ECs, and their effects on EC phenotype such as sprouting angiogenesis.
Collapse
Affiliation(s)
- Raid Alhayaza
- Alfaisal University School of Medicine, Riyadh, Saudi Arabia
| | - Emaan Haque
- Alfaisal University School of Medicine, Riyadh, Saudi Arabia
| | - Catherine Karbasiafshar
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| |
Collapse
|
39
|
Notch Signaling Function in the Angiocrine Regulation of Tumor Development. Cells 2020; 9:cells9112467. [PMID: 33198378 PMCID: PMC7697556 DOI: 10.3390/cells9112467] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022] Open
Abstract
The concept of tumor growth being angiogenesis dependent had its origin in the observations of Judah Folkman in 1969 of a retinoblastoma in a child. Tumor angiogenesis is initiated when endothelial cells (ECs) respond to local stimuli and migrate towards the growing mass, which results in the formation of tubular structures surrounded by perivascular support cells that transport blood to the inner tumor. In turn, the neo-vasculature supports tumor development and eventual metastasis. This process is highly regulated by several signaling pathways. Central to this process is the Notch signaling pathway. Beyond the role of Notch signaling in tumor angiogenesis, a major hallmark of cancer development, it has also been implicated in the regulation of tumor cell proliferation and survival, in epithelial-to-mesenchymal transition, invasion and metastasis and in the regulation of cancer stem cells, in a variety of hematologic and solid malignancies. There is increasing evidence for the tumor vasculature being important in roles other than those linked to blood perfusion. Namely, endothelial cells act on and influence neighboring tumor cells by use of angiocrine factors to generate a unique cellular microenvironment, thereby regulating tumor stem-like cells’ homeostasis, modulating tumor progression, invasiveness, trafficking and metastasis. This review will focus on Notch signaling components that play a part in angiocrine signaling in a tumor setting.
Collapse
|
40
|
Palano MT, Giannandrea D, Platonova N, Gaudenzi G, Falleni M, Tosi D, Lesma E, Citro V, Colombo M, Saltarella I, Ria R, Amodio N, Taiana E, Neri A, Vitale G, Chiaramonte R. Jagged Ligands Enhance the Pro-Angiogenic Activity of Multiple Myeloma Cells. Cancers (Basel) 2020; 12:cancers12092600. [PMID: 32932949 PMCID: PMC7565520 DOI: 10.3390/cancers12092600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 12/27/2022] Open
Abstract
Simple Summary The Jagged family of ligands are aberrantly expressed during multiple myeloma progression and contributes to activate Notch signaling both in myeloma cells and in the nearby bone marrow cell populations activating several pro-tumor effects. This work elucidates, in vitro, in vivo as well as in patients’ bone marrow biopsies, different mechanisms by which tumor cell-derived Jagged1 and 2 contribute to myeloma-associated angiogenesis. These include the ability to induce myeloma and bone marrow stromal cell secretion of VEGF along with a direct activation of the pro-angiogenic Notch signaling pathway in endothelial cells. This research provides a rational for a Jagged-directed therapy in multiple myeloma. Abstract Multiple myeloma (MM) is an incurable plasma cell malignancy arising primarily within the bone marrow (BM). During MM progression, different modifications occur in the tumor cells and BM microenvironment, including the angiogenic shift characterized by the increased capability of endothelial cells to organize a network, migrate and express angiogenic factors, including vascular endothelial growth factor (VEGF). Here, we studied the functional outcome of the dysregulation of Notch ligands, Jagged1 and Jagged2, occurring during disease progression, on the angiogenic potential of MM cells and BM stromal cells (BMSCs). Jagged1–2 expression was modulated by RNA interference or soluble peptide administration, and the effects on the MM cell lines’ ability to induce human pulmonary artery cells (HPAECs) angiogenesis or to indirectly increase the BMSC angiogenic potential was analyzed in vitro; in vivo validation was performed on a zebrafish model and MM patients’ BM biopsies. Overall, our results indicate that the MM-derived Jagged ligands (1) increase the tumor cell angiogenic potential by directly triggering Notch activation in the HPAECs or stimulating the release of angiogenic factors, i.e., VEGF; and (2) stimulate the BMSCs to promote angiogenesis through VEGF secretion. The observed pro-angiogenic effect of Notch activation in the BM during MM progression provides further evidence of the potential of a therapy targeting the Jagged ligands.
Collapse
Affiliation(s)
- Maria Teresa Palano
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (M.T.P.); (D.G.); (N.P.); (M.F.); (D.T.); (E.L.); (V.C.); (M.C.)
| | - Domenica Giannandrea
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (M.T.P.); (D.G.); (N.P.); (M.F.); (D.T.); (E.L.); (V.C.); (M.C.)
| | - Natalia Platonova
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (M.T.P.); (D.G.); (N.P.); (M.F.); (D.T.); (E.L.); (V.C.); (M.C.)
| | - Germano Gaudenzi
- Istituto Auxologico Italiano, IRCCS, Laboratory of Geriatric and Oncologic Neuroendocrinology Research, 20095 Cusano Milanino, Italy; (G.G.); (G.V.)
| | - Monica Falleni
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (M.T.P.); (D.G.); (N.P.); (M.F.); (D.T.); (E.L.); (V.C.); (M.C.)
| | - Delfina Tosi
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (M.T.P.); (D.G.); (N.P.); (M.F.); (D.T.); (E.L.); (V.C.); (M.C.)
| | - Elena Lesma
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (M.T.P.); (D.G.); (N.P.); (M.F.); (D.T.); (E.L.); (V.C.); (M.C.)
| | - Valentina Citro
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (M.T.P.); (D.G.); (N.P.); (M.F.); (D.T.); (E.L.); (V.C.); (M.C.)
| | - Michela Colombo
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (M.T.P.); (D.G.); (N.P.); (M.F.); (D.T.); (E.L.); (V.C.); (M.C.)
| | - Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine and Clinical Oncology, University of Bari Medical School, 70124 Bari, Italy; (I.S.); (R.R.)
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine and Clinical Oncology, University of Bari Medical School, 70124 Bari, Italy; (I.S.); (R.R.)
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Elisa Taiana
- Department of Oncology and Hemato-Oncology, University of Milano. Hematology, Fondazione Ca’ Granda IRCCS Policlinico, 20122 Milano, Italy; (E.T.); (A.N.)
| | - Antonino Neri
- Department of Oncology and Hemato-Oncology, University of Milano. Hematology, Fondazione Ca’ Granda IRCCS Policlinico, 20122 Milano, Italy; (E.T.); (A.N.)
| | - Giovanni Vitale
- Istituto Auxologico Italiano, IRCCS, Laboratory of Geriatric and Oncologic Neuroendocrinology Research, 20095 Cusano Milanino, Italy; (G.G.); (G.V.)
- Department of Clinical Sciences and Community Health (DISCCO), University of Milan, 20122 Milan, Italy
| | - Raffaella Chiaramonte
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (M.T.P.); (D.G.); (N.P.); (M.F.); (D.T.); (E.L.); (V.C.); (M.C.)
- Correspondence: ; Tel.: +39-02-50323249
| |
Collapse
|
41
|
Endothelial Jagged1 Antagonizes Dll4/Notch Signaling in Decidual Angiogenesis during Early Mouse Pregnancy. Int J Mol Sci 2020; 21:ijms21186477. [PMID: 32899448 PMCID: PMC7554752 DOI: 10.3390/ijms21186477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 12/22/2022] Open
Abstract
Maternal spiral arteries and newly formed decidual capillaries support embryonic development prior to placentation. Previous studies demonstrated that Notch signaling is active in endothelial cells of both decidual capillaries and spiral arteries, however the role of Notch signaling in physiologic decidual angiogenesis and maintenance of the decidual vasculature in early mouse pregnancy has not yet been fully elucidated. We used the Cdh5-CreERT2;Jagged1(Jag1)flox/flox (Jag1∆EC) mouse model to delete Notch ligand, Jag1, in maternal endothelial cells during post-implantation, pre-placentation mouse pregnancy. Loss of endothelial Jag1 leads to increased expression of Notch effectors, Hey2 and Nrarp, and increased endothelial Notch signaling activity in areas of the decidua with remodeling angiogenesis. This correlated with an increase in Dll4 expression in capillary endothelial cells, but not spiral artery endothelial cells. Consistent with increased Dll4/Notch signaling, we observed decreased VEGFR2 expression and endothelial cell proliferation in angiogenic decidual capillaries. Despite aberrant Dll4 expression and Notch activation in Jag1∆EC mutants, pregnancies were maintained and the decidual vasculature was not altered up to embryonic day 7.5. Thus, Jag1 functions in the newly formed decidual capillaries as an antagonist of endothelial Dll4/Notch signaling during angiogenesis, but Jag1 signaling is not necessary for early uterine angiogenesis.
Collapse
|
42
|
The oncogenic role of Jagged1/Notch signaling in cancer. Biomed Pharmacother 2020; 129:110416. [PMID: 32593969 DOI: 10.1016/j.biopha.2020.110416] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/14/2022] Open
Abstract
Aberrant activation of Notch signaling plays an oncogenic role in cancer development. Jagged1 (JAG1) is an important Notch ligand that triggers Notch signaling through cell-cell interactions. JAG1 overexpression has been reported in many different types of cancer and correlates with a poor clinical prognosis. JAG1/Notch signaling controls oncogenic processes in different cell types and cellular contexts. Furthermore, JAG1/Notch signaling cascades activate a number of oncogenic factors that regulate cellular functions such as proliferation, metastasis, drug-resistance, and angiogenesis. To suppress the severe toxicity of pan-Notch inhibitors, JAG1 is attracting increasing attention as a source of therapeutic targets for cancers. In this review, the oncogenic role of JAG1/Notch signaling in cancer is discussed, as well as implications of strategies to inhibit JAG1/Notch signaling activity.
Collapse
|
43
|
Minami T, Muramatsu M, Kume T. Organ/Tissue-Specific Vascular Endothelial Cell Heterogeneity in Health and Disease. Biol Pharm Bull 2020; 42:1609-1619. [PMID: 31582649 DOI: 10.1248/bpb.b19-00531] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The vascular system forms the largest surface in our body, serving as a critical interface between blood circulation and our diverse organ/tissue environments. Thus, the vascular system performs a gatekeeper function for organ/tissue homeostasis and the body's adjustment to pathological challenges. The endothelium, as the most inner layer of the vasculature, regulates the tissue microenvironment, which is critical for development, hemostatic balance, inflammation, and angiogenesis, with a role as well in tumor malignancy and metastasis. These multitudinous functions are primarily mediated by organ/tissue-specifically differentiated endothelial cells, in which heterogeneity has long been recognized at the molecular and histological level. Based on these general principles of vascular-bed heterogeneity and characterization, this review largely covers landmark discoveries regarding organ/tissue microenvironment-governed endothelial cell phenotypic changes. These involve the physical features of continuous, discontinuous, fenestrated, and sinusoidal endothelial cells, in addition to the more specialized endothelial cell layers of the lymphatic system, glomerulus, tumors, and the blood brain barrier (BBB). Major signal pathways of endothelial specification are outlined, including Notch as a key factor of tip/stalk- and arterial-endothelial cell differentiation. We also denote the shear stress sensing machinery used to convey blood flow-mediated biophysical forces that are indispensable to maintaining inert and mature endothelial phenotypes. Since our circulatory system is among the most fundamental and emergent targets of study in pharmacology from the viewpoint of drug metabolism and delivery, a better molecular understanding of organ vasculature-bed heterogeneity may lead to better strategies for novel vascular-targeted treatments to fight against hitherto intractable diseases.
Collapse
Affiliation(s)
- Takashi Minami
- Div. of Molecular and Vascular Biology, IRDA, Kumamoto University
| | | | - Tsutomu Kume
- Div. of Molecular and Vascular Biology, IRDA, Kumamoto University.,Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine
| |
Collapse
|
44
|
Notch Signaling and Embryonic Development: An Ancient Friend, Revisited. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:9-37. [PMID: 32060869 DOI: 10.1007/978-3-030-34436-8_2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The evolutionary highly conserved Notch pathway, which first developed during evolution in metazoans and was first discovered in fruit flies (Drosophila melanogaster), governs many core processes including cell fate decisions during embryonic development. A huge mountain of scientific evidence convincingly demonstrates that Notch signaling represents one of the most important pathways that regulate embryogenesis from sponges, roundworms, Drosophila melanogaster, and mice to humans. In this review, we give a brief introduction on how Notch orchestrates the embryonic development of several selected tissues, summarizing some of the most relevant findings in the central nervous system, skin, kidneys, liver, pancreas, inner ear, eye, skeleton, heart, and vascular system.
Collapse
|
45
|
Malashicheva A, Kostina A, Kostareva A, Irtyuga O, Gordeev M, Uspensky V. Notch signaling in the pathogenesis of thoracic aortic aneurysms: A bridge between embryonic and adult states. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165631. [PMID: 31816439 DOI: 10.1016/j.bbadis.2019.165631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/23/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022]
Abstract
Aneurysms of the thoracic aorta are a "silent killer" with no evident clinical signs until the fatal outcome. Molecular and genetic bases of thoracic aortic aneurysms mainly include transforming growth factor beta signaling, smooth muscle contractile units and metabolism genes, and extracellular matrix genes. In recent studies, a role of Notch signaling, among other pathways, has emerged in disease pathogenesis. Notch is a highly conserved signaling pathway that regulates the development and differentiation of many types of tissues and influences major cellular processes such as cell proliferation, differentiation and apoptosis. Mutations in several Notch signaling components have been associated with a number of heart defects, demonstrating an essential role of Notch signaling both in cardiovascular system development and its maintenance during postnatal life. This review discusses the role of Notch signaling in the pathogenesis of thoracic aortic aneurysms considering development and maintenance of the aortic root and how developmental regulations by Notch signaling may influence thoracic aortic aneurysms.
Collapse
Affiliation(s)
- Anna Malashicheva
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia; Institute of Cytology, Russian Academy of Sciences, Tikhoretskiy, 4, 194064 Saint Petersburg, Russia; Saint Petersburg State University, Department of Embryology, Universitetskaya nab., 7/9, 199034, Saint Petersburg, Russia.
| | - Aleksandra Kostina
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia; Institute of Cytology, Russian Academy of Sciences, Tikhoretskiy, 4, 194064 Saint Petersburg, Russia
| | - Anna Kostareva
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| | - Olga Irtyuga
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| | - Mikhail Gordeev
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| | - Vladimir Uspensky
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| |
Collapse
|
46
|
Travisano SI, Oliveira VL, Prados B, Grego-Bessa J, Piñeiro-Sabarís R, Bou V, Gómez MJ, Sánchez-Cabo F, MacGrogan D, de la Pompa JL. Coronary arterial development is regulated by a Dll4-Jag1-EphrinB2 signaling cascade. eLife 2019; 8:49977. [PMID: 31789590 PMCID: PMC6917494 DOI: 10.7554/elife.49977] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 12/01/2019] [Indexed: 12/29/2022] Open
Abstract
Coronaries are essential for myocardial growth and heart function. Notch is crucial for mouse embryonic angiogenesis, but its role in coronary development remains uncertain. We show Jag1, Dll4 and activated Notch1 receptor expression in sinus venosus (SV) endocardium. Endocardial Jag1 removal blocks SV capillary sprouting, while Dll4 inactivation stimulates excessive capillary growth, suggesting that ligand antagonism regulates coronary primary plexus formation. Later endothelial ligand removal, or forced expression of Dll4 or the glycosyltransferase Mfng, blocks coronary plexus remodeling, arterial differentiation, and perivascular cell maturation. Endocardial deletion of Efnb2 phenocopies the coronary arterial defects of Notch mutants. Angiogenic rescue experiments in ventricular explants, or in primary human endothelial cells, indicate that EphrinB2 is a critical effector of antagonistic Dll4 and Jag1 functions in arterial morphogenesis. Thus, coronary arterial precursors are specified in the SV prior to primary coronary plexus formation and subsequent arterial differentiation depends on a Dll4-Jag1-EphrinB2 signaling cascade.
Collapse
Affiliation(s)
- Stanislao Igor Travisano
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Vera Lucia Oliveira
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Belén Prados
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Joaquim Grego-Bessa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Rebeca Piñeiro-Sabarís
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Vanesa Bou
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Manuel J Gómez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| |
Collapse
|
47
|
Cho SJ, Yun SM, Jo C, Jeong J, Park MH, Han C, Koh YH. Altered expression of Notch1 in Alzheimer's disease. PLoS One 2019; 14:e0224941. [PMID: 31770379 PMCID: PMC6879159 DOI: 10.1371/journal.pone.0224941] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/19/2019] [Indexed: 02/07/2023] Open
Abstract
Notch signaling is an evolutionarily conserved pathway that regulates cell-cell interactions through binding of Notch family receptors to their cognate ligands. Notch signaling has an essential role in vascular development and angiogenesis. Recent studies have reported that Notch may be implicated in Alzheimer's disease (AD) pathophysiology. We measured the levels of soluble Notch1 (sNotch1) in the plasma samples from 72 dementia patients (average age 75.1 y), 89 subjects with amnestic mild cognitive impairment (MCI) (average age 73.72 y), and 150 cognitively normal controls (average age 72.34 y). Plasma levels of sNotch1 were 25.27% lower in dementia patients as compared to healthy control subjects. However, the level of Notch1 protein was significantly increased in human brain microvascular endothelial cells (HBMECs) after amyloid-beta treatment. Also, Notch1 mRNA level was significantly increased in HBMECs and iPSC-derived neuronal cells from AD patient compared to normal control. These results indicate that altered expression of Notch1 might be associated with the risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Sun-Jung Cho
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Sang-Moon Yun
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Chulman Jo
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Jihyun Jeong
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Moon Ho Park
- Departments of Neurology, Korea University Medical College, Ansan Hospital, 123 Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do, Republic of Korea
| | - Changsu Han
- Departments of Psychiatry, Korea University Medical College, Ansan Hospital, 123 Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do, Republic of Korea
| | - Young Ho Koh
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
48
|
HIF-1α is Overexpressed in Odontogenic Keratocyst Suggesting Activation of HIF-1α and NOTCH1 Signaling Pathways. Cells 2019; 8:cells8070731. [PMID: 31319505 PMCID: PMC6678339 DOI: 10.3390/cells8070731] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022] Open
Abstract
Background: The odontogenic keratocyst (OKC) is an odontogenic cyst that shows aggressive and intriguing biological behavior. It is suggested that a hypoxic environment occurs in OKC, which led us to investigate the immunoexpression and location of hypoxia-inducible factor 1-alpha (HIF-1α) and other hypoxia-related proteins. Methods: Twenty cases of OKC were evaluated for the expression of Notch homolog 1 (NOTCH1), HIF-1α, disintegrin and metalloproteinase domain-containing protein 12 (ADAM-12), and heparin-binding epidermal growth factor-like growth factor (HBEGF) by immunohistochemistry and compared to eight control cases of calcifying odontogenic cystic (COC), orthokeratinized odontogenic cyst (OOC), and normal oral mucosa (OM) in basal and parabasal layers. Results: In OKC, all the proteins tested were expressed significantly higher in both basal (except for NOTCH1 and HBEGF in OOC) and suprabasal epithelial layers compared to controls. Looking at the epithelial layers within OKC, we observed an increased NOTCH1 and HIF-1α expression in parabasal layers. Conclusions: These results suggest that hypoxia occurs more intensively in OKC compared to COC, OM, and OOC. Hypoxia appeared to be stronger in parabasal layers as observed by higher HIF-1α expression in upper cells. Overexpression of NOTCH1, ADAM-12, and HBEGF in OKC was observed, which suggests that microenvironmental hypoxia could potentially regulate the expression of hypoxia-related proteins, and consequently, its clinical and biological behavior.
Collapse
|
49
|
Aquila G, Kostina A, Vieceli Dalla Sega F, Shlyakhto E, Kostareva A, Marracino L, Ferrari R, Rizzo P, Malaschicheva A. The Notch pathway: a novel therapeutic target for cardiovascular diseases? Expert Opin Ther Targets 2019; 23:695-710. [PMID: 31304807 DOI: 10.1080/14728222.2019.1641198] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The Notch pathway is involved in determining cell fate during development and postnatally in continuously renewing tissues, such as the endothelium, the epithelium, and in the stem cells pool. The dysregulation of the Notch pathway is one of the causes of limited response, or resistance, to available cancer treatments and novel therapeutic strategies based on Notch inhibition are being investigated in preclinical and clinical studies in oncology. A large body of evidence now shows that the dysregulation of the Notch pathway is also involved in the pathophysiology of cardiovascular diseases (CVDs). Areas covered: This review discusses the molecular mechanisms involving Notch which underlie heart failure, aortic valve calcification, and aortic aneurysm. Expert opinion: Despite the existence of preventive, pharmacological and surgical interventions approaches, CVDs are the first causes of mortality worldwide. The Notch pathway is becoming increasingly recognized as being involved in heart failure, aortic aneurysm and aortic valve calcification, which are among the most common global causes of mortality due to CVDs. As already shown in cancer, the dissection of the biological processes and molecular mechanisms involving Notch should pave the way for new strategies to prevent and cure these diseases.
Collapse
Affiliation(s)
- Giorgio Aquila
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Aleksandra Kostina
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia.,Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences , St-Petersburg , Russia
| | | | - Eugeniy Shlyakhto
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia
| | - Anna Kostareva
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia
| | - Luisa Marracino
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy
| | - Roberto Ferrari
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy.,Maria Cecilia Hospital, GVM Care & Research , Cotignola , Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research , Cotignola , Italy.,Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy
| | - Anna Malaschicheva
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia.,Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences , St-Petersburg , Russia.,Department of Embryology, Faculty of Biology, Saint-Petersburg State University , St. Petersburg , Russia
| |
Collapse
|
50
|
Zhang L, Zhang X, Xu H, Huang L, Zhang S, Liu W, Yang Y, Fei P, Li S, Yang M, Zhao P, Zhu X, Yang Z. Exome sequencing revealed Notch ligand JAG1 as a novel candidate gene for familial exudative vitreoretinopathy. Genet Med 2019; 22:77-84. [DOI: 10.1038/s41436-019-0571-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/28/2019] [Indexed: 01/12/2023] Open
|