1
|
Moghassemi S, Nikanfar S, Dadashzadeh A, Sousa MJ, Wan Y, Sun F, Colson A, De Windt S, Kwaspen L, Kanbar M, Sobhani K, Yang J, Vlieghe H, Li Y, Debiève F, Wyns C, Amorim CA. The revolutionary role of placental derivatives in biomedical research. Bioact Mater 2025; 49:456-485. [PMID: 40177109 PMCID: PMC11964572 DOI: 10.1016/j.bioactmat.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The human placenta is a transient yet crucial organ that plays a key role in sustaining the relationship between the maternal and fetal organisms. Despite its historical classification as "biowaste," placental tissues have garnered increasing attention since the early 1900s for their significant medical potential, particularly in wound repair and surgical application. As ethical considerations regarding human placental derivatives have largely been assuaged in many countries, they have gained significant attention due to their versatile applications in various biomedical fields, such as biomedical engineering, regenerative medicine, and pharmacology. Moreover, there is a substantial trend toward various animal product substitutions in laboratory research with human placental derivatives, reflecting a broader commitment to advancing ethical and sustainable research methodologies. This review provides a comprehensive examination of the current applications of human placental derivatives, explores the mechanisms behind their therapeutic effects, and outlines the future potential and directions of this rapidly advancing field.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Saba Nikanfar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yuting Wan
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Fengxuan Sun
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Arthur Colson
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sven De Windt
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Lena Kwaspen
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Keyvan Sobhani
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yongqian Li
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christine Wyns
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
2
|
Samatoshenkov IV, Aimaletdinov AM, Zakirova EY, Kalmykov EL, Khodzhibaev R, Samatoshenkova YM, Ganiev IM, Kadyrov MS, Mukhamedshina YO. Extended Toxicity, Genotoxicity, and Mutagenicity of Combination of pBudK-coVEGF-coANG and pBudK-coGDNF Plasmids in Preclinical Trials. Biomedicines 2025; 13:1223. [PMID: 40427050 DOI: 10.3390/biomedicines13051223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Chronic lower limb ischemia is a debilitating condition, particularly prevalent among elderly patients and individuals ineligible for revascularization procedures. Gene therapy aimed at promoting therapeutic angiogenesis presents a promising alternative treatment strategy. Objectives: This study evaluated the preclinical safety of a gene therapy drug composed of the plasmids pBudK-coVEGF-coANG and pBudK-coGDNF in laboratory animals. Safety assessment followed a single intramuscular injection at a dose 30 times higher than the proposed therapeutic level. Methods: Acute toxicity was monitored over a 24-h period. Genotoxicity was assessed using the micronucleus test at doses of 200, 1000, and 5000 μg/kg. Bone marrow cytology was analyzed to detect hematopoietic toxicity. Delayed toxicity was evaluated over a two-week recovery period. Results: No signs of acute toxicity were observed, even at the highest dose. The micronucleus test revealed no genotoxic effects, with no significant increase in micronucleated polychromatic erythrocytes compared to control groups. Bone marrow erythroblast parameters remained within normal physiological ranges. Additionally, no delayed adverse effects were detected during the recovery period. Conclusions: The gene therapy drug demonstrated a favorable preclinical safety profile, exhibiting no evidence of toxicity or genotoxicity, even at substantially elevated doses. These findings support the continued development of this therapy as a potential treatment for chronic lower limb ischemia in patients who are not candidates for surgical intervention.
Collapse
Affiliation(s)
- Igor V Samatoshenkov
- Medizinische Hochschule Brandenburg Theodor Fontane, 14770 Brandenburg, Germany
- Department of Vascular and Endovascular Surgery, Brandenburg University Clinic, 14770 Brandenburg, Germany
| | - Alexander M Aimaletdinov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Elena Y Zakirova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Egan L Kalmykov
- Department of Vascular and Endovascular Surgery, Brandenburg University Clinic, 14770 Brandenburg, Germany
| | - Rustam Khodzhibaev
- Department of Vascular and Endovascular Surgery, Brandenburg University Clinic, 14770 Brandenburg, Germany
| | - Yulia M Samatoshenkova
- SAHI Republican Clinical Hospital of the Ministry of Health of the Republic of Tatarstan, 420138 Kazan, Russia
| | - Ilnur M Ganiev
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | | | - Yana O Mukhamedshina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
- Department of Histology, Cytology and Embryology, Kazan State Medical University, 420012 Kazan, Russia
- Academy of Sciences of the Republic of Tatarstan, 420013 Kazan, Russia
| |
Collapse
|
3
|
Li J, Luo J, Wang T, Tian X, Xu C, Wang W, Zhang D. DNA methylation associated with the serum alanine aminotransferase concentration: evidence from Chinese monozygotic twins. Clin Epigenetics 2025; 17:65. [PMID: 40296130 PMCID: PMC12039056 DOI: 10.1186/s13148-025-01869-1] [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: 12/04/2024] [Accepted: 03/30/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND To identify nongenetic factors influences on DNA methylation (DNAm) variations associated with blood Alanine Aminotransferase (ALT) concentration, this study conducted an epigenome-wide association study (EWAS) on Chinese monozygotic twins. METHODS A total of 61 pairs of Chinese monozygotic twins involved in this study. Whole blood samples were analyzed for DNAm profiling using the Reduced Representation Bisulfite Sequencing (RRBS) technique. We examined the relationship between DNAm levels at each CpG site and serum ALT using a linear mixed-effects model. Enrichment analysis and causal inference analysis was conducted, and differentially methylated regions (DMRs) were further identified. Candidate CpGs were validated in a community sample. Genome-wide significance were calculated by Bonferroni correction (p < 2.14 × 10-7). RESULTS We identified 85 CpGs reaching genome-wide significance (p < 2.14 × 10-7), located in 16 genes including FLT4, ADARB2, MRPS31P2, and RELB. Causal inference suggested that DNAm at 61 out of 85 significant CpGs within 14 genes influenced ALT level. 52 DMRs and 1765 pathways such as low voltage-gated calcium channel activity and focal adhesion were identified having influences on ALT levels. Further validation using community population found four CpGs mapped to FLT4 and three to RELB showing hypomethylation and hypermethylation in cases with abnormal ALT (ALT > 40 U/L), respectively. CONCLUSION This study identified several differentially methylated CpG sites associated with serum ALT in the Chinese population, particularly within FLT4 and RELB. These findings provide new insights into the epigenetic modifications underlying liver function.
Collapse
Affiliation(s)
- Jingxian Li
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China
| | - Jia Luo
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China
| | - Tong Wang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China
| | - Xiaocao Tian
- Qingdao Municipal Centre for Disease Control and Prevention, No.175 Shandong Road, Qingdao, 266033, Shandong Province, People's Republic of China
| | - Chunsheng Xu
- Qingdao Municipal Centre for Disease Control and Prevention, No.175 Shandong Road, Qingdao, 266033, Shandong Province, People's Republic of China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China.
| |
Collapse
|
4
|
Lin YY, Warren E, Macklin BL, Ramirez L, Gerecht S. Endothelial-pericyte interactions regulate angiogenesis via VEGFR2 signaling during retinal development and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.08.642174. [PMID: 40161680 PMCID: PMC11952325 DOI: 10.1101/2025.03.08.642174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Pericytes stabilize the microvasculature by enhancing endothelial barrier integrity, resulting in functional networks. During retinal development, pericyte recruitment is crucial for stabilizing nascent angiogenic vasculature. However, in adulthood, disrupted endothelial-pericyte interactions lead to vascular dropout and pathological angiogenesis in ocular microvascular diseases, and strategies to stabilize the retinal vasculature are lacking. We demonstrate that direct endothelial-pericyte contact downregulates pVEGFR2 in endothelial cells, which enhances pericyte migration and promotes endothelial cell barrier function. Intravitreal injection of a VEGFR2 inhibitor in mouse models of the developing retina and oxygen-induced retinopathy increased pericyte recruitment and aided vascular stability. The VEGFR2 inhibitor further rescued ischemic retinopathy by enhancing vascularization and tissue growth while reducing vascular permeability. Our findings offer a druggable target to support the growth of functional and mature microvasculature in ocular microvascular diseases and tissue regeneration overall.
Collapse
|
5
|
Haupt HB, Nickerson JM, Boatright JH, Pardue MT, Bales KL. Effects of treadmill exercise on retinal vascular morphology, function, and circulating immune factors in a mouse model of retinal degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637694. [PMID: 39990328 PMCID: PMC11844490 DOI: 10.1101/2025.02.11.637694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Purpose Exercise is neuroprotective in rodents undergoing retinal degeneration (RD). However, the effects of exercise on retinal vasculature remain unexplored. Here, we investigate whether treadmill exercise influences retinal vascular morphology, function, gene expression, and circulating factors in a light-induced retinal degeneration (LIRD) mouse model. Methods 6-week-old female BALB/c mice were assigned to inactive+dim, active+dim, inactive+LIRD and active+LIRD groups (n=20 per group). Active mice were treadmill exercised (1hr/d 10m/min) for two weeks, then LIRD was induced (5000 lux/4hrs). Inactive mice were placed on a static treadmill. Retinal neurovascular coupling was measured with functional hyperemia (FH) and vascular morphology using OCT-A. Vascular gene expression was quantified from isolated retinal endothelial cells using ddPCR five days following LIRD. Serum was collected for circulating cytokine and chemokine analyses. Data were analyzed using 2-way ANOVA. Results Retinal vessel vasodilation was significantly increased in active+LIRD mice compared to inactive+LIRD mice. Superficial and intermediate/deep vascular plexi from inactive+LIRD mice had significantly decreased vessel density and total vessel length, with increased numbers of end points and lacunarity compared to active groups. Isolated retinal endothelial cell gene expression varied among groups. Most notably, Active+LIRD mice had a distinct immune response profile, with increased expression of IL-6, KC, and VEGF-A. Conclusions Treadmill exercise maintained retinal vascular morphology and function, modestly altered endothelial gene expression, and is associated with a specific circulating immune response profile in a LIRD mouse model. These data indicate therapeutic effects of exercise on retinal vasculature in RD.
Collapse
Affiliation(s)
- Hayden B Haupt
- Atlanta VA Medical Center for Visual and Neurocognitive Rehabilitation, Decatur, GA
| | | | - Jeffrey H Boatright
- Atlanta VA Medical Center for Visual and Neurocognitive Rehabilitation, Decatur, GA
- Department of Ophthalmology, Emory University, Atlanta, GA
| | - Machelle T Pardue
- Atlanta VA Medical Center for Visual and Neurocognitive Rehabilitation, Decatur, GA
- Department of Ophthalmology, Emory University, Atlanta, GA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Katie L Bales
- Atlanta VA Medical Center for Visual and Neurocognitive Rehabilitation, Decatur, GA
| |
Collapse
|
6
|
Sun Y, Liu J, Chen K, Zhong N, He C, Luan X, Zang X, Sun J, Cao N, Wang W, Ren Q. Polydopamine grafting polyether ether ketone to stabilize growth factor for efficient osteonecrosis repair. Sci Rep 2025; 15:3697. [PMID: 39880837 PMCID: PMC11779900 DOI: 10.1038/s41598-025-86965-1] [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: 07/29/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
This study examines the biocompatibility, osteogenic potential, and effectiveness of polyether ether ketone (PEEK) composites for treating osteonecrosis, seeking to establish a theoretical basis for clinical application. A range of PEEK composite materials, including sulfonated polyether ether ketone (SPEEK), polydopamine-sulfonated polyether ether ketone (SPEEK-PDA), bone-forming peptide-poly-dopamine-sulfonated polyether ether ketone (SPEEK-PDA-BFP), and vascular endothelial growth factor-poly-dopamine-sulfonated polyether ether ketone (SPEEK-PDA-VEGF), were constructed by concentrated sulfuric acid sulfonation, polydopamine modification and grafting of bioactive factors. The experiments involved adult male New Zealand rabbits aged 24-28 weeks and weighing 2.6-4 kg. The SPEEK-PDA-BFP possesses the smallest water contact angle, indicating the highest hydrophilicity, with its surface characterized by a rich density of clustered BFP particles. The SPEEK-PDA-BFP exhibits superior adhesion, proliferation, and differentiation capabilities, along with pronounced bacteriostatic effects, which are attributed to its dense particle clusters. The SPEEK-PDA-BFP facilitates the formation of regular and dense bone trabeculae. Comparative study on treating osteonecrosis with SPEEK-PDA-VEGF and SPEEK-PDA-BFP highlighted the superior formation of mature bone trabeculae and angiogenic protein CD31 around SPEEK-PDA-VEGF. The PEEK composite materials have good biocompatibility, osteogenic activity and bone repair activity. In particular, SPEEK-PDA-VEGF composite materials have the best effect on bone repair.
Collapse
Affiliation(s)
- Yi Sun
- Department of Bone Joint, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256600, China
| | - Jingyun Liu
- Department of Orthopedics, Liaocheng People's Hospital, No. 67, Dongchang West Road, Liaocheng, 252000, China
| | - Kaijia Chen
- Department of Bone Joint, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256600, China
| | - Nannan Zhong
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Chengpeng He
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Xinming Luan
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Xiaobei Zang
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Jianbo Sun
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Ning Cao
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Wenbo Wang
- Department of Orthopedics, The Second Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan, 250001, China
| | - Qiang Ren
- Department of Bone Joint, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256600, China.
| |
Collapse
|
7
|
Raudales A, Schager B, Hancock D, Narayana K, Sharma S, Reeson P, Oshanyk A, Cheema M, Körbelin J, Brown CE. Angiogenesis in the mature mouse cortex is governed in a regional- and Notch1-dependent manner. Cell Rep 2024; 43:115029. [PMID: 39612246 DOI: 10.1016/j.celrep.2024.115029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/10/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
Cerebral angiogenesis is well appreciated in development and after injury, but the extent to which it occurs across cortical regions in normal adult mice and the underlying mechanisms are incompletely understood. Using in vivo imaging, we show that angiogenesis in anterior-medial cortical regions (retrosplenial and sensorimotor cortex) was exceptionally rare. By contrast, angiogenesis was significantly elevated in posterior-lateral regions such as visual cortex, primarily within 200 μm of the cortical surface. There was no effect of sex on angiogenesis rates, nor were there regional differences in vessel pruning (for either sex). To understand the mechanisms, we surveyed gene expression and found that Notch-related genes were enriched in ultra-stable retrosplenial cortex. Using endothelial-specific knockdown of Notch1, cerebral angiogenesis was significantly increased along with genes implicated in angiogenesis (Apln, Angpt2, Cdkn1a). Our study shows that angiogenesis is regionally dependent and that manipulations of Notch1 could unlock the angiogenic potential of the mature vasculature.
Collapse
Affiliation(s)
- Alejandra Raudales
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Ben Schager
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Dominique Hancock
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Kamal Narayana
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Sorabh Sharma
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Patrick Reeson
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Adam Oshanyk
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Manjinder Cheema
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
8
|
Perez-Gutierrez L, Li P, Ferrara N. Endothelial cell diversity: the many facets of the crystal. FEBS J 2024; 291:3287-3302. [PMID: 36266750 DOI: 10.1111/febs.16660] [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: 07/17/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Endothelial cells (ECs) form the inner lining of blood vessels and play crucial roles in angiogenesis. While it has been known for a long time that there are considerable differences among ECs from lymphatic and blood vessels, as well as among arteries, veins and capillaries, the full repertoire of endothelial diversity is only beginning to be elucidated. It has become apparent that the role of ECs is not just limited to their exchange functions. Indeed, a multitude of organ-specific functions, including release of growth factors, regulation of immune functions, have been linked to ECs. Recent years have seen a surge into the identification of spatiotemporal molecular and functional heterogeneity of ECs, supported by technologies such as single-cell RNA sequencing (scRNA-seq), lineage tracing and intersectional genetics. Together, these techniques have spurred the generation of epigenomic, transcriptomic and proteomic signatures of ECs. It is now clear that ECs across organs and in different vascular beds, but even within the same vessel, have unique molecular identities and employ specialized molecular mechanisms to fulfil highly specialized needs. Here, we focus on the molecular heterogeneity of the endothelium in different organs and pathological conditions.
Collapse
Affiliation(s)
- Lorena Perez-Gutierrez
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Pin Li
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| |
Collapse
|
9
|
McDermott JG, Goodlett BL, Creed HA, Navaneethabalakrishnan S, Rutkowski JM, Mitchell BM. Inflammatory Alterations to Renal Lymphatic Endothelial Cell Gene Expression in Mouse Models of Hypertension. Kidney Blood Press Res 2024; 49:588-604. [PMID: 38972305 PMCID: PMC11345939 DOI: 10.1159/000539721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 06/02/2024] [Indexed: 07/09/2024] Open
Abstract
INTRODUCTION Hypertension (HTN) is a major cardiovascular disease that can cause and be worsened by renal damage and inflammation. We previously reported that renal lymphatic endothelial cells (LECs) increase in response to HTN and that augmenting lymphangiogenesis in the kidneys reduces blood pressure and renal pro-inflammatory immune cells in mice with various forms of HTN. Our aim was to evaluate the specific changes that renal LECs undergo in HTN. METHODS We performed single-cell RNA sequencing. Using the angiotensin II-induced and salt-sensitive mouse models of HTN, we isolated renal CD31+ and podoplanin+ cells. RESULTS Sequencing of these cells revealed three distinct cell types with unique expression profiles, including LECs. The number and transcriptional diversity of LECs increased in samples from mice with HTN, as demonstrated by 597 differentially expressed genes (p < 0.01), 274 significantly enriched pathways (p < 0.01), and 331 regulons with specific enrichment in HTN LECs. These changes demonstrate a profound inflammatory response in renal LECs in HTN, leading to an increase in genes and pathways associated with inflammation-driven growth and immune checkpoint activity in LECs. CONCLUSION These results reinforce and help to further explain the benefits of renal LECs and lymphangiogenesis in HTN.
Collapse
Affiliation(s)
- Justin G. McDermott
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | - Bethany L. Goodlett
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | - Heidi A. Creed
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | | | - Joseph M. Rutkowski
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | - Brett M. Mitchell
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| |
Collapse
|
10
|
Garcia-Gonzalez I, Rocha SF, Hamidi A, Garcia-Ortega L, Regano A, Sanchez-Muñoz M, Lytvyn M, Garcia-Cabero A, Roig-Soucase S, Schoofs H, Castro M, Sabata H, Potente M, Graupera M, Makinen T, Benedito R. iSuRe-HadCre is an essential tool for effective conditional genetics. Nucleic Acids Res 2024; 52:e56. [PMID: 38850155 PMCID: PMC11260470 DOI: 10.1093/nar/gkae472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/04/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024] Open
Abstract
Methods for modifying gene function at high spatiotemporal resolution in mice have revolutionized biomedical research, with Cre-loxP being the most widely used technology. However, the Cre-loxP technology has several drawbacks, including weak activity, leakiness, toxicity, and low reliability of existing Cre-reporters. This is mainly because different genes flanked by loxP sites (floxed) vary widely in their sensitivity to Cre-mediated recombination. Here, we report the generation, validation, and utility of iSuRe-HadCre, a new dual Cre-reporter and deleter mouse line that avoids these drawbacks. iSuRe-HadCre achieves this through a novel inducible dual-recombinase genetic cascade that ensures that cells expressing a fluorescent reporter had only transient Cre activity, that is nonetheless sufficient to effectively delete floxed genes. iSuRe-HadCre worked reliably in all cell types and for the 13 floxed genes tested. This new tool will enable the precise, efficient, and trustworthy analysis of gene function in entire mouse tissues or in single cells.
Collapse
Affiliation(s)
- Irene Garcia-Gonzalez
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Susana F Rocha
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Anahita Hamidi
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Lourdes Garcia-Ortega
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alvaro Regano
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Maria S Sanchez-Muñoz
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mariya Lytvyn
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Aroa Garcia-Cabero
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sergi Roig-Soucase
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Hans Schoofs
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Marco Castro
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Helena Sabata
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mariona Graupera
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
- ICREA, Institució Catalana de Recerca i Estudis Avançats, Pg. Lluís Companys 23, Barcelona, Spain
| | - Taija Makinen
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
- Translational Cancer Medicine Program, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
- Wihuri Research Institute, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
11
|
Sultan I, Ramste M, Peletier P, Hemanthakumar KA, Ramanujam D, Tirronen A, von Wright Y, Antila S, Saharinen P, Eklund L, Mervaala E, Ylä-Herttuala S, Engelhardt S, Kivelä R, Alitalo K. Contribution of VEGF-B-Induced Endocardial Endothelial Cell Lineage in Physiological Versus Pathological Cardiac Hypertrophy. Circ Res 2024; 134:1465-1482. [PMID: 38655691 PMCID: PMC11542978 DOI: 10.1161/circresaha.123.324136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/19/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Preclinical studies have shown the therapeutic potential of VEGF-B (vascular endothelial growth factor B) in revascularization of the ischemic myocardium, but the associated cardiac hypertrophy and adverse side effects remain a concern. To understand the importance of endothelial proliferation and migration for the beneficial versus adverse effects of VEGF-B in the heart, we explored the cardiac effects of autocrine versus paracrine VEGF-B expression in transgenic and gene-transduced mice. METHODS We used single-cell RNA sequencing to compare cardiac endothelial gene expression in VEGF-B transgenic mouse models. Lineage tracing was used to identify the origin of a VEGF-B-induced novel endothelial cell population and adeno-associated virus-mediated gene delivery to compare the effects of VEGF-B isoforms. Cardiac function was investigated using echocardiography, magnetic resonance imaging, and micro-computed tomography. RESULTS Unlike in physiological cardiac hypertrophy driven by a cardiomyocyte-specific VEGF-B transgene (myosin heavy chain alpha-VEGF-B), autocrine VEGF-B expression in cardiac endothelium (aP2 [adipocyte protein 2]-VEGF-B) was associated with septal defects and failure to increase perfused subendocardial capillaries postnatally. Paracrine VEGF-B led to robust proliferation and myocardial migration of a novel cardiac endothelial cell lineage (VEGF-B-induced endothelial cells) of endocardial origin, whereas autocrine VEGF-B increased proliferation of VEGF-B-induced endothelial cells but failed to promote their migration and efficient contribution to myocardial capillaries. The surviving aP2-VEGF-B offspring showed an altered ratio of secreted VEGF-B isoforms and developed massive pathological cardiac hypertrophy with a distinct cardiac vessel pattern. In the normal heart, we found a small VEGF-B-induced endothelial cell population that was only minimally expanded during myocardial infarction but not during physiological cardiac hypertrophy associated with mouse pregnancy. CONCLUSIONS Paracrine and autocrine secretions of VEGF-B induce expansion of a specific endocardium-derived endothelial cell population with distinct angiogenic markers. However, autocrine VEGF-B signaling fails to promote VEGF-B-induced endothelial cell migration and contribution to myocardial capillaries, predisposing to septal defects and inducing a mismatch between angiogenesis and myocardial growth, which results in pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Ibrahim Sultan
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Markus Ramste
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Pim Peletier
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Karthik Amudhala Hemanthakumar
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technical University of Munich, DZHK partner site Munich Heart Alliance, Germany (D.R., S.E.)
- RNATICS GmbH, Planegg, Germany (D.R.)
| | - Annakaisa Tirronen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (A.T., S.Y.-H.)
| | - Ylva von Wright
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Salli Antila
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Pipsa Saharinen
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Finland (L.E.)
| | - Eero Mervaala
- Department of Pharmacology (E.M.), Faculty of Medicine, University of Helsinki, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (A.T., S.Y.-H.)
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich, DZHK partner site Munich Heart Alliance, Germany (D.R., S.E.)
| | - Riikka Kivelä
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Stem Cells and Metabolism Research Program (R.K.), Faculty of Medicine, University of Helsinki, Finland
- Faculty of Sport and Health Sciences, University of Jyväskylä, Finland (R.K.)
| | - Kari Alitalo
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| |
Collapse
|
12
|
Wang J, Yan YL, Yu XY, Pan JY, Liu XL, Hong LL, Wang B. Meroterpenoids from Marine Sponge Hyrtios sp. and Their Anticancer Activity against Human Colorectal Cancer Cells. Mar Drugs 2024; 22:183. [PMID: 38667800 PMCID: PMC11051118 DOI: 10.3390/md22040183] [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: 04/08/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Two new meroterpenoids, hyrtamide A (1) and hyrfarnediol A (2), along with two known ones, 3-farnesyl-4-hydroxybenzoic acid methyl ester (3) and dictyoceratin C (4), were isolated from a South China Sea sponge Hyrtios sp. Their structures were elucidated by NMR and MS data. Compounds 2-4 exhibited weak cytotoxicity against human colorectal cancer cells (HCT-116), showing IC50 values of 41.6, 45.0, and 37.3 μM, respectively. Furthermore, compounds 3 and 4 significantly suppressed the invasion of HCT-116 cells while also downregulating the expression of vascular endothelial growth factor receptor 1 (VEGFR-1) and vimentin proteins, which are key markers associated with angiogenesis and epithelial-mesenchymal transition (EMT). Our findings suggest that compounds 3 and 4 may exert their anti-invasive effects on tumor cells by inhibiting the expression of VEGFR-1 and impeding the process of EMT.
Collapse
Affiliation(s)
- Jie Wang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Yue-Lu Yan
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Xin-Yi Yu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Jia-Yan Pan
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Xin-Lian Liu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Li-Li Hong
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bin Wang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| |
Collapse
|
13
|
Xue J, Deng J, Qin H, Yan S, Zhao Z, Qin L, Liu J, Wang H. The interaction of platelet-related factors with tumor cells promotes tumor metastasis. J Transl Med 2024; 22:371. [PMID: 38637802 PMCID: PMC11025228 DOI: 10.1186/s12967-024-05126-6] [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: 01/16/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Platelets not only participate in thrombosis and hemostasis but also interact with tumor cells and protect them from mechanical damage caused by hemodynamic shear stress and natural killer cell lysis, thereby promoting their colonization and metastasis to distant organs. Platelets can affect the tumor microenvironment via interactions between platelet-related factors and tumor cells. Metastasis is a key event in cancer-related death and is associated with platelet-related factors in lung, breast, and colorectal cancers. Although the factors that promote platelet expression vary slightly in terms of their type and mode of action, they all contribute to the overall process. Recognizing the correlation and mechanisms between these factors is crucial for studying the colonization of distant target organs and developing targeted therapies for these three types of tumors. This paper reviews studies on major platelet-related factors closely associated with metastasis in lung, breast, and colorectal cancers.
Collapse
Affiliation(s)
- Jie Xue
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
- Department of Blood Transfusion, The Central Hospital of Qingdao Jiaozhou, 99 Yunxi River South Road, Qingdao, 266300, Shandong, China
| | - Jianzhao Deng
- Clinical Laboratory, The Central Hospital of Qingdao Jiaozhou, 99 Yunxi River South Road, Qingdao, 266300, Shandong, China
| | - Hongwei Qin
- Department of Blood Transfusion, The Central Hospital of Qingdao Jiaozhou, 99 Yunxi River South Road, Qingdao, 266300, Shandong, China
| | - Songxia Yan
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Zhen Zhao
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Lifeng Qin
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Jiao Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Haiyan Wang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China.
| |
Collapse
|
14
|
Lang L, Liang S, Zhang F, Fu Y, Wang J, Deng K, Wang L, Gao P, Zhu C, Shu G, Wu R, Jiang Q, Wang S. Knockdown of the VEGFB/VEGFR1 signaling suppresses pubertal mammary gland development of mice via the inhibition of PI3K/Akt pathway. Int J Biol Macromol 2024; 264:130782. [PMID: 38471613 DOI: 10.1016/j.ijbiomac.2024.130782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Vascular endothelial growth factor B (VEGFB) has been well demonstrated to play a crucial role in regulating vascular function by binding to the VEGF receptors (VEGFRs). However, the specific role of VEGFB and VEGFRs in pubertal mammary gland development remains unclear. In this study, we observed that blocking the VEGF receptors with Axitinib suppressed the pubertal mammary gland development. Meanwhile, the proliferation of mammary epithelial cells (HC11) was repressed by blocking the VEGF receptors with Axitinib. Additionally, knockdown of VEGFR1 rather than VEGFR2 and NRP1 elicited the inhibition of HC11 proliferation, suggesting the essential role of VEGFR1 during this process. Furthermore, Axitinib or VEGFR1 knockdown led to the inhibition of the PI3K/Akt pathway. However, the inhibition of HC11 proliferation induced by Axitinib and or VEGFR1 knockdown was eliminated by the Akt activator SC79, indicating the involvement of the PI3K/Akt pathway. Finally, the knockdown of VEGFB and VEGFR1 suppressed the pubertal development of mice mammary gland with the inhibition of the PI3K/Akt pathway. In summary, the results showed that knockdown of the VEGFB/VEGFR1 signaling suppresses pubertal mammary gland development of mice via the inhibition of the PI3K/Akt pathway, which provides a new target for the regulation of pubertal mammary gland development.
Collapse
Affiliation(s)
- Limin Lang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Shuyi Liang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Fenglin Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Yiming Fu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Junfeng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Kaixin Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Canjun Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Ruifan Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China; Yunfu Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Research Institute of Wens Foodstuff Group Co., Ltd., Xinxing 527400, PR China.
| |
Collapse
|
15
|
Osinski V, Yellamilli A, Firulyova MM, Zhang MJ, Peck A, Auger JL, Faragher JL, Marath A, Voeller RK, O’Connell TD, Zaitsev K, Binstadt BA. Profibrotic VEGFR3-Dependent Lymphatic Vessel Growth in Autoimmune Valvular Carditis. Arterioscler Thromb Vasc Biol 2024; 44:807-821. [PMID: 38269589 PMCID: PMC10978259 DOI: 10.1161/atvbaha.123.320326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Rheumatic heart disease is the major cause of valvular heart disease in developing nations. Endothelial cells (ECs) are considered crucial contributors to rheumatic heart disease, but greater insight into their roles in disease progression is needed. METHODS We used a Cdh5-driven EC lineage-tracing approach to identify and track ECs in the K/B.g7 model of autoimmune valvular carditis. Single-cell RNA sequencing was used to characterize the EC populations in control and inflamed mitral valves. Immunostaining and conventional histology were used to evaluate lineage tracing and validate single-cell RNA-sequencing findings. The effects of VEGFR3 (vascular endothelial growth factor receptor 3) and VEGF-C (vascular endothelial growth factor C) inhibitors were tested in vivo. The functional impact of mitral valve disease in the K/B.g7 mouse was evaluated using echocardiography. Finally, to translate our findings, we analyzed valves from human patients with rheumatic heart disease undergoing mitral valve replacements. RESULTS Lineage tracing in K/B.g7 mice revealed new capillary lymphatic vessels arising from valve surface ECs during the progression of disease in K/B.g7 mice. Unsupervised clustering of mitral valve single-cell RNA-sequencing data revealed novel lymphatic valve ECs that express a transcriptional profile distinct from other valve EC populations including the recently identified PROX1 (Prospero homeobox protein 1)+ lymphatic valve ECs. During disease progression, these newly identified lymphatic valve ECs expand and upregulate a profibrotic transcriptional profile. Inhibiting VEGFR3 through multiple approaches prevented expansion of this mitral valve lymphatic network. Echocardiography demonstrated that K/B.g7 mice have left ventricular dysfunction and mitral valve stenosis. Valve lymphatic density increased with age in K/B.g7 mice and correlated with worsened ventricular dysfunction. Importantly, human rheumatic valves contained similar lymphatics in greater numbers than nonrheumatic controls. CONCLUSIONS These studies reveal a novel mode of inflammation-associated, VEGFR3-dependent postnatal lymphangiogenesis in murine autoimmune valvular carditis, with similarities to human rheumatic heart disease.
Collapse
Affiliation(s)
- Victoria Osinski
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Amritha Yellamilli
- Department of Pediatrics, Stanford School of Medicine, Palo Alto, CA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN
| | - Maria M. Firulyova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Michael J. Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Alyssa Peck
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jennifer L. Auger
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jessica L. Faragher
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | | | | | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| | - Konstantin Zaitsev
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Bryce A. Binstadt
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
16
|
Liu J, Yan S, Du J, Teng L, Yang R, Xu P, Tao W. Mechanism and treatment of diarrhea associated with tyrosine kinase inhibitors. Heliyon 2024; 10:e27531. [PMID: 38501021 PMCID: PMC10945189 DOI: 10.1016/j.heliyon.2024.e27531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/20/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have become first-line drugs for cancer treatment. However, their clinical use is seriously hindered since many patients experience diarrhea after receiving TKIs. The mechanisms of TKI-associated diarrhea remain unclear. Most existing therapies are symptomatic treatments based on experience and their effects are unsatisfactory. Therefore, clarification of the mechanisms underlying diarrhea is critical to develop effective anti-diarrhea drugs. This article summarizes several potential mechanisms of TKI-associated diarrhea and reviews current treatment progress.
Collapse
Affiliation(s)
- Jiangnan Liu
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, 150001, PR China
- Key Laboratory of Acoustic, Optical and Electromagnetic Diagnosis and Treatment of Cardiovascular Diseases, Heilongjiang, 150001, PR China
- The Cell Transplantation Key Laboratory of National Health Commission, Heilongjiang, 150001, PR China
| | - Shuai Yan
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, 150001, PR China
- Key Laboratory of Acoustic, Optical and Electromagnetic Diagnosis and Treatment of Cardiovascular Diseases, Heilongjiang, 150001, PR China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, 150001, PR China
- The Cell Transplantation Key Laboratory of National Health Commission, Heilongjiang, 150001, PR China
| | - Juntong Du
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, 150001, PR China
- Key Laboratory of Acoustic, Optical and Electromagnetic Diagnosis and Treatment of Cardiovascular Diseases, Heilongjiang, 150001, PR China
- The Cell Transplantation Key Laboratory of National Health Commission, Heilongjiang, 150001, PR China
| | - Lizhi Teng
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, 150001, PR China
- Key Laboratory of Acoustic, Optical and Electromagnetic Diagnosis and Treatment of Cardiovascular Diseases, Heilongjiang, 150001, PR China
- The Cell Transplantation Key Laboratory of National Health Commission, Heilongjiang, 150001, PR China
| | - Ru Yang
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, 150001, PR China
- Key Laboratory of Acoustic, Optical and Electromagnetic Diagnosis and Treatment of Cardiovascular Diseases, Heilongjiang, 150001, PR China
- The Cell Transplantation Key Laboratory of National Health Commission, Heilongjiang, 150001, PR China
| | - Peng Xu
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, 150001, PR China
- Key Laboratory of Acoustic, Optical and Electromagnetic Diagnosis and Treatment of Cardiovascular Diseases, Heilongjiang, 150001, PR China
- The Cell Transplantation Key Laboratory of National Health Commission, Heilongjiang, 150001, PR China
| | - Weiyang Tao
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, 150001, PR China
- Key Laboratory of Acoustic, Optical and Electromagnetic Diagnosis and Treatment of Cardiovascular Diseases, Heilongjiang, 150001, PR China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang, 150001, PR China
- The Cell Transplantation Key Laboratory of National Health Commission, Heilongjiang, 150001, PR China
| |
Collapse
|
17
|
Li DS, Liao HX, Zhang JL, Qin B. Effect of aflibercept combined with triamcinolone acetonide on aqueous humor growth factor and inflammatory mediators in diabetic macular edema. Int J Ophthalmol 2024; 17:297-303. [PMID: 38371257 PMCID: PMC10827616 DOI: 10.18240/ijo.2024.02.11] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/28/2023] [Indexed: 02/20/2024] Open
Abstract
AIM To investigate the efficacy of aflibercept combined with sub-tenon injection of triamcinolone acetonide (TA) in treating diabetic macular edema (DME) and to examine changes in growth factors and inflammatory mediator levels in aqueous humor after injection. METHODS Totally 67 DME patients (67 eyes) and 30 cataract patients (32 eyes) were enrolled as the DME group and the control group, respectively. The DME group was divided into the aflibercept group (34 cases) and the aflibercept combined with TA group (combined group, 33 cases). The aqueous humor of both groups was collected during the study period. The aqueous levels of vascular endothelial growth factor (VEGF), monocyte chemoattractant protein-1 (MCP-1), interleukin-6 (IL-6), interleukin-8 (IL-8), and interleukin-1β (IL-1β) were detected using a microsphere suspension array technology (Luminex 200TM). Aqueous cytokines, best-corrected visual acuity (BCVA), central macular thickness (CMT), and complications before and after treatment were compared between the aflibercept group and combined group. RESULTS The concentrations of VEGF, MCP-1, IL-6, and IL-8 in the aqueous humor were significantly higher in the DME group than those of the control group (all P<0.01). After 1mo of surgery, the concentrations of VEGF, MCP-1, IL-6, and IL-8 in the aqueous humor were significantly lower in the combined group than those of the aflibercept group (all P<0.01). The BCVA and CMT values of the two groups were statistically different after 1 and 2mo of treatment (P<0.01). However, the difference was not statistically significant after 3mo of treatment (P>0.05). CONCLUSION The cytokines VEGF, MCP-1, IL-6, and IL-8 in the aqueous humor of DME patients are significantly increased. Aflibercept and aflibercept combined with TA have good efficacy in DME patients, can effectively reduce CMT, improve the patient's vision, and have high safety. Aflibercept combined with TA can quickly down-regulate the aqueous humor cytokines and help to relieve macular edema rapidly. However, the long-term efficacy is comparable to that of aflibercept alone.
Collapse
Affiliation(s)
- De-Shuang Li
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen 518031, Guangdong Province, China
| | - Hong-Xia Liao
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen 518031, Guangdong Province, China
| | - Jing-Lin Zhang
- Guangzhou Aier Eye Hospital, Jinan University, Guangzhou 510040, Guangdong Province, China
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen 518031, Guangdong Province, China
| |
Collapse
|
18
|
Sarabipour S, Kinghorn K, Quigley KM, Kovacs-Kasa A, Annex BH, Bautch VL, Mac Gabhann F. Trafficking dynamics of VEGFR1, VEGFR2, and NRP1 in human endothelial cells. PLoS Comput Biol 2024; 20:e1011798. [PMID: 38324585 PMCID: PMC10878527 DOI: 10.1371/journal.pcbi.1011798] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 02/20/2024] [Accepted: 01/03/2024] [Indexed: 02/09/2024] Open
Abstract
The vascular endothelial growth factor (VEGF) family of cytokines are key drivers of blood vessel growth and remodeling. These ligands act via multiple VEGF receptors (VEGFR) and co-receptors such as Neuropilin (NRP) expressed on endothelial cells. These membrane-associated receptors are not solely expressed on the cell surface, they move between the surface and intracellular locations, where they can function differently. The location of the receptor alters its ability to 'see' (access and bind to) its ligands, which regulates receptor activation; location also alters receptor exposure to subcellularly localized phosphatases, which regulates its deactivation. Thus, receptors in different subcellular locations initiate different signaling, both in terms of quantity and quality. Similarly, the local levels of co-expression of other receptors alters competition for ligands. Subcellular localization is controlled by intracellular trafficking processes, which thus control VEGFR activity; therefore, to understand VEGFR activity, we must understand receptor trafficking. Here, for the first time, we simultaneously quantify the trafficking of VEGFR1, VEGFR2, and NRP1 on the same cells-specifically human umbilical vein endothelial cells (HUVECs). We build a computational model describing the expression, interaction, and trafficking of these receptors, and use it to simulate cell culture experiments. We use new quantitative experimental data to parameterize the model, which then provides mechanistic insight into the trafficking and localization of this receptor network. We show that VEGFR2 and NRP1 trafficking is not the same on HUVECs as on non-human ECs; and we show that VEGFR1 trafficking is not the same as VEGFR2 trafficking, but rather is faster in both internalization and recycling. As a consequence, the VEGF receptors are not evenly distributed between the cell surface and intracellular locations, with a very low percentage of VEGFR1 being on the cell surface, and high levels of NRP1 on the cell surface. Our findings have implications both for the sensing of extracellular ligands and for the composition of signaling complexes at the cell surface versus inside the cell.
Collapse
Affiliation(s)
- Sarvenaz Sarabipour
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Karina Kinghorn
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kaitlyn M. Quigley
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Anita Kovacs-Kasa
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, United States of America
| | - Brian H. Annex
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, United States of America
| | - Victoria L. Bautch
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Feilim Mac Gabhann
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
19
|
Wendong Y, Jiali J, Qiaomei F, Yayun W, Xianze X, Zheng S, Wei H. Biomechanical forces and force-triggered drug delivery in tumor neovascularization. Biomed Pharmacother 2024; 171:116117. [PMID: 38171243 DOI: 10.1016/j.biopha.2023.116117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024] Open
Abstract
Tumor angiogenesis is one of the typical hallmarks of tumor occurrence and development, and tumor neovascularization also exhibits distinct characteristics from normal blood vessels. As the number of cells and matrix inside the tumor increases, the biomechanical force is enhanced, specifically manifested as solid stress, fluid stress, stiffness, and topology. This mechanical microenvironment also provides shelter for tumors and intensifies angiogenesis, providing oxygen and nutritional support for tumor progression. During tumor development, the biomechanical microenvironment also emerges, which in turn feeds back to regulate the tumor progression, including tumor angiogenesis, and biochemical and biomechanical signals can regulate tumor angiogenesis. Blood vessels possess inherent sensitivity to mechanical stimuli, but compared to the extensive research on biochemical signal regulation, the study of the regulation of tumor neovascularization by biomechanical signals remains relatively scarce. Biomechanical forces can affect the phenotypic characteristics and mechanical signaling pathways of tumor blood vessels, directly regulating angiogenesis. Meanwhile, they can indirectly regulate tumor angiogenesis by causing an imbalance in angiogenesis signals and affecting stromal cell function. Understanding the regulatory mechanism of biomechanical forces in tumor angiogenesis is beneficial for better identifying and even taming the mechanical forces involved in angiogenesis, providing new therapeutic targets for tumor vascular normalization. Therefore, we summarized the composition of biomechanical forces and their direct or indirect regulation of tumor neovascularization. In addition, this review discussed the use of biomechanical forces in combination with anti-angiogenic therapies for the treatment of tumors, and biomechanical forces triggered delivery systems.
Collapse
Affiliation(s)
- Yao Wendong
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Jiang Jiali
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Fan Qiaomei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Weng Yayun
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Xie Xianze
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Shi Zheng
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| | - Huang Wei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| |
Collapse
|
20
|
Wang L, Liu WQ, Broussy S, Han B, Fang H. Recent advances of anti-angiogenic inhibitors targeting VEGF/VEGFR axis. Front Pharmacol 2024; 14:1307860. [PMID: 38239196 PMCID: PMC10794590 DOI: 10.3389/fphar.2023.1307860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Vascular endothelial growth factors (VEGF), Vascular endothelial growth factor receptors (VEGFR) and their downstream signaling pathways are promising targets in anti-angiogenic therapy. They constitute a crucial system to regulate physiological and pathological angiogenesis. In the last 20 years, many anti-angiogenic drugs have been developed based on VEGF/VEGFR system to treat diverse cancers and retinopathies, and new drugs with improved properties continue to emerge at a fast rate. They consist of different molecular structures and characteristics, which enable them to inhibit the interaction of VEGF/VEGFR, to inhibit the activity of VEGFR tyrosine kinase (TK), or to inhibit VEGFR downstream signaling. In this paper, we reviewed the development of marketed anti-angiogenic drugs involved in the VEGF/VEGFR axis, as well as some important drug candidates in clinical trials. We discuss their mode of action, their clinical benefits, and the current challenges that will need to be addressed by the next-generation of anti-angiogenic drugs. We focus on the molecular structures and characteristics of each drug, including those approved only in China.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, Zhejiang Xiaoshan Hospital, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Wang-Qing Liu
- CiTCoM, CNRS, INSERM, Université Paris Cité, Paris, France
| | | | - Bingnan Han
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Hongming Fang
- Department of Oncology, Zhejiang Xiaoshan Hospital, Hangzhou, China
| |
Collapse
|
21
|
Bernier-Latmani J, González-Loyola A, Petrova TV. Mechanisms and functions of intestinal vascular specialization. J Exp Med 2024; 221:e20222008. [PMID: 38051275 PMCID: PMC10697212 DOI: 10.1084/jem.20222008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
The intestinal vasculature has been studied for the last 100 years, and its essential role in absorbing and distributing ingested nutrients is well known. Recently, fascinating new insights into the organization, molecular mechanisms, and functions of intestinal vessels have emerged. These include maintenance of intestinal epithelial cell function, coping with microbiota-induced inflammatory pressure, recruiting gut-specific immune cells, and crosstalk with other organs. Intestinal function is also regulated at the systemic and cellular levels, such that the postprandial hyperemic response can direct up to 30% of systemic blood to gut vessels, while micron-sized endothelial cell fenestrations are necessary for nutrient uptake. In this review, we will highlight past discoveries made about intestinal vasculature in the context of new findings of molecular mechanisms underpinning gut function. Such comprehensive understanding of the system will pave the way to breakthroughs in nutrient uptake optimization, drug delivery efficiency, and treatment of human diseases.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | | | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
22
|
Li J, Li XL, Li CQ. Immunoregulation mechanism of VEGF signaling pathway inhibitors and its efficacy on the kidney. Am J Med Sci 2023; 366:404-412. [PMID: 37699444 DOI: 10.1016/j.amjms.2023.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 05/10/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023]
Abstract
Angiogenesis and immunosuppression are closely related pathophysiologic processes. Widely prescribed in malignant tumor and proliferative retinal lesions, VEGF signaling pathway inhibitors may cause hypertension and renal injury in some patients, presenting with proteinuria, nephrotic syndrome, renal failure and thrombotic microangiopathy. VEGF signaling pathway inhibitors block the action of both VEGF-A and VEGF-C. However, VEGF-A and VEGF-C produced by podocytes are vital to maintain the physiological function of glomerular endothelial cells and podocytes. There is still no effective treatment for kidney disease associated with VEGF signaling pathway inhibitors and some patients have progressive renal failure even after withdrawal of the drug. Recent studies reveal that blocking of VEGF-A and VEGF-C can activate CD4 +and CD8+ T cells, augment antigen-presenting function of dendritic cells, enhance cytotoxicity of macrophages and initiate complement cascade activation. VEGF and VEGFR are expressed in immune cells, which are involved in the immunosuppression and cross-talk among immune cells. This review summarizes the expression and function of VEGF-A and VEGF-C in the kidney. The current immunoregulation mechanisms of VEGF signaling pathway inhibitors are reviewed. Finally, combinate strategies are summarized to highlight the proposal for VEGF signaling pathway inhibitors.
Collapse
Affiliation(s)
- Jun Li
- Department of Nephrology, Affiliated Hospital of Jiangnan University, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Jiangsu, China.
| | - Xiao-Lin Li
- Wuxi School of Medicine, Jiangnan University, Jiangsu, China
| | - Chun-Qing Li
- Department of Nephrology, Affiliated Hospital of Jiangnan University, Jiangsu, China
| |
Collapse
|
23
|
Pérez-Gutiérrez L, Ferrara N. Biology and therapeutic targeting of vascular endothelial growth factor A. Nat Rev Mol Cell Biol 2023; 24:816-834. [PMID: 37491579 DOI: 10.1038/s41580-023-00631-w] [Citation(s) in RCA: 149] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 07/27/2023]
Abstract
The formation of new blood vessels, called angiogenesis, is an essential pathophysiological process in which several families of regulators have been implicated. Among these, vascular endothelial growth factor A (VEGFA; also known as VEGF) and its two tyrosine kinase receptors, VEGFR1 and VEGFR2, represent a key signalling pathway mediating physiological angiogenesis and are also major therapeutic targets. VEGFA is a member of the gene family that includes VEGFB, VEGFC, VEGFD and placental growth factor (PLGF). Three decades after its initial isolation and cloning, VEGFA is arguably the most extensively investigated signalling system in angiogenesis. Although many mediators of angiogenesis have been identified, including members of the FGF family, angiopoietins, TGFβ and sphingosine 1-phosphate, all current FDA-approved anti-angiogenic drugs target the VEGF pathway. Anti-VEGF agents are widely used in oncology and, in combination with chemotherapy or immunotherapy, are now the standard of care in multiple malignancies. Anti-VEGF drugs have also revolutionized the treatment of neovascular eye disorders such as age-related macular degeneration and ischaemic retinal disorders. In this Review, we emphasize the molecular, structural and cellular basis of VEGFA action as well as recent findings illustrating unexpected interactions with other pathways and provocative reports on the role of VEGFA in regenerative medicine. We also discuss clinical and translational aspects of VEGFA. Given the crucial role that VEGFA plays in regulating angiogenesis in health and disease, this molecule is largely the focus of this Review.
Collapse
Affiliation(s)
- Lorena Pérez-Gutiérrez
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
24
|
Sjöberg E, Melssen M, Richards M, Ding Y, Chanoca C, Chen D, Nwadozi E, Pal S, Love DT, Ninchoji T, Shibuya M, Simons M, Dimberg A, Claesson-Welsh L. Endothelial VEGFR2-PLCγ signaling regulates vascular permeability and antitumor immunity through eNOS/Src. J Clin Invest 2023; 133:e161366. [PMID: 37651195 PMCID: PMC10575733 DOI: 10.1172/jci161366] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/15/2023] [Indexed: 09/02/2023] Open
Abstract
Endothelial phospholipase Cγ (PLCγ) is essential for vascular development; however, its role in healthy, mature, or pathological vessels is unexplored. Here, we show that PLCγ was prominently expressed in vessels of several human cancer forms, notably in renal cell carcinoma (RCC). High PLCγ expression in clear cell RCC correlated with angiogenic activity and poor prognosis, while low expression correlated with immune cell activation. PLCγ was induced downstream of vascular endothelial growth factor receptor 2 (VEGFR2) phosphosite Y1173 (pY1173). Heterozygous Vegfr2Y1173F/+ mice or mice lacking endothelial PLCγ (Plcg1iECKO) exhibited a stabilized endothelial barrier and diminished vascular leakage. Barrier stabilization was accompanied by decreased expression of immunosuppressive cytokines, reduced infiltration of B cells, helper T cells and regulatory T cells, and improved response to chemo- and immunotherapy. Mechanistically, pY1173/PLCγ signaling induced Ca2+/protein kinase C-dependent activation of endothelial nitric oxide synthase (eNOS), required for tyrosine nitration and activation of Src. Src-induced phosphorylation of VE-cadherin at Y685 was accompanied by disintegration of endothelial junctions. This pY1173/PLCγ/eNOS/Src pathway was detected in both healthy and tumor vessels in Vegfr2Y1173F/+ mice, which displayed decreased activation of PLCγ and eNOS and suppressed vascular leakage. Thus, we believe that we have identified a clinically relevant endothelial PLCγ pathway downstream of VEGFR2 pY1173, which destabilizes the endothelial barrier and results in loss of antitumor immunity.
Collapse
Affiliation(s)
- Elin Sjöberg
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Marit Melssen
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Mark Richards
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Yindi Ding
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Catarina Chanoca
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Dongying Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emmanuel Nwadozi
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Sagnik Pal
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Dominic T. Love
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Takeshi Ninchoji
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Masabumi Shibuya
- Institute of Physiology and Medicine, Jobu University, Takasaki, Gunma, Japan
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| |
Collapse
|
25
|
Ge Z, Zhang Q, Lin W, Jiang X, Zhang Y. The role of angiogenic growth factors in the immune microenvironment of glioma. Front Oncol 2023; 13:1254694. [PMID: 37790751 PMCID: PMC10542410 DOI: 10.3389/fonc.2023.1254694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Angiogenic growth factors (AGFs) are a class of secreted cytokines related to angiogenesis that mainly include vascular endothelial growth factors (VEGFs), stromal-derived factor-1 (SDF-1), platelet-derived growth factors (PDGFs), fibroblast growth factors (FGFs), transforming growth factor-beta (TGF-β) and angiopoietins (ANGs). Accumulating evidence indicates that the role of AGFs is not only limited to tumor angiogenesis but also participating in tumor progression by other mechanisms that go beyond their angiogenic role. AGFs were shown to be upregulated in the glioma microenvironment characterized by extensive angiogenesis and high immunosuppression. AGFs produced by tumor and stromal cells can exert an immunomodulatory role in the glioma microenvironment by interacting with immune cells. This review aims to sum up the interactions among AGFs, immune cells and cancer cells with a particular emphasis on glioma and tries to provide new perspectives for understanding the glioma immune microenvironment and in-depth explorations for anti-glioma therapy.
Collapse
Affiliation(s)
| | | | | | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yanyu Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
26
|
Mao Y, Ge H, Chen W, Wang Y, Liu H, Li Z, Bai Y, Wang D, Yu Y, Zhen Q, Li B, Sun L. RasGRP1 influences imiquimod-induced psoriatic inflammation via T-cell activation in mice. Int Immunopharmacol 2023; 122:110590. [PMID: 37429143 DOI: 10.1016/j.intimp.2023.110590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/12/2023]
Abstract
The vascular endothelial growth factor (VEGF) signal transduction pathway has been shown to be a potential target for the treatment of psoriasis. Ras guanyl-releasing protein 1 (RasGRP1), a downstream target gene of VEGF, regulates the development, homeostasis, and differentiation of T cells, but the contribution of RasGRP1 to psoriasis is limited. In this manuscript, we aimed to investigate the role of RasGRP1 in psoriasis. The RNA-Seq transcriptome sequencing data from the mouse model of psoriasis treated with IMQ (imiquimod) were analyzed. The effect of RasGRP1 was investigated through in vivo injection of activators or small molecular inhibitors, as well as adeno-associated virus injections. Gene knockout and NB-UVB (narrow-band ultraviolet B) treatments were utilized to interfere with the psoriatic mouse model. By transfection of lentivirus in vitro, the effect of RasGRP1 gene function on the secretion of psoriasis-related cytokines by T cells was confirmed. We showed that cutaneous VEGF and RasGRP1 were strongly activated in human psoriatic lesions and the skin of mice with IMQ-induced psoriasis. RasGRP1 deficiency and overexpression influence IMQ-induced psoriasis-like manifestations and skin inflammation in mice. VEGF, secreted mainly by epidermal cells, mediates psoriatic inflammation through the RasGRP1-AKT-NF-κB pathway. RasGRP1 is required for psoriasis development mediated by VEGF. These results confirmed the role of RasGRP1 in the pathogenesis of psoriasis and provided potential targets for clinical psoriasis treatment.
Collapse
Affiliation(s)
- Yiwen Mao
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Huiyao Ge
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Weiwei Chen
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - YiRui Wang
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Hao Liu
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Zhuo Li
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Yuanming Bai
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Daiyue Wang
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Yafen Yu
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Qi Zhen
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Bao Li
- Integrated Laboratory, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Liangdan Sun
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Health Science Center, North China University of Science and Technology, Tangshan 063210, China; North China University of Science and Technology Affiliated Hospital, Tangshan 063000, China; Inflammation and Immune Diseases Laboratory of North China University of Science and Technology, Tangshan 063210, China; School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| |
Collapse
|
27
|
Kam CY, Singh ID, Gonzalez DG, Matte-Martone C, Solá P, Solanas G, Bonjoch J, Marsh E, Hirschi KK, Greco V. Mechanisms of skin vascular maturation and maintenance captured by longitudinal imaging of live mice. Cell 2023; 186:2345-2360.e16. [PMID: 37167971 PMCID: PMC10225355 DOI: 10.1016/j.cell.2023.04.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/03/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023]
Abstract
A functional network of blood vessels is essential for organ growth and homeostasis, yet how the vasculature matures and maintains homeostasis remains elusive in live mice. By longitudinally tracking the same neonatal endothelial cells (ECs) over days to weeks, we found that capillary plexus expansion is driven by vessel regression to optimize network perfusion. Neonatal ECs rearrange positions to evenly distribute throughout the developing plexus and become positionally stable in adulthood. Upon local ablation, adult ECs survive through a plasmalemmal self-repair response, while neonatal ECs are predisposed to die. Furthermore, adult ECs reactivate migration to assist vessel repair. Global ablation reveals coordinated maintenance of the adult vascular architecture that allows for eventual network recovery. Lastly, neonatal remodeling and adult maintenance of the skin vascular plexus are orchestrated by temporally restricted, neonatal VEGFR2 signaling. Our work sheds light on fundamental mechanisms that underlie both vascular maturation and adult homeostasis in vivo.
Collapse
Affiliation(s)
- Chen Yuan Kam
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ishani D Singh
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - David G Gonzalez
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Paloma Solá
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Guiomar Solanas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Júlia Bonjoch
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Edward Marsh
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Cell Biology and Dermatology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
28
|
Ghalehbandi S, Yuzugulen J, Pranjol MZI, Pourgholami MH. The role of VEGF in cancer-induced angiogenesis and research progress of drugs targeting VEGF. Eur J Pharmacol 2023; 949:175586. [PMID: 36906141 DOI: 10.1016/j.ejphar.2023.175586] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/16/2023] [Accepted: 02/08/2023] [Indexed: 03/11/2023]
Abstract
Angiogenesis is a double-edged sword; it is a mechanism that defines the boundary between health and disease. In spite of its central role in physiological homeostasis, it provides the oxygen and nutrition needed by tumor cells to proceed from dormancy if pro-angiogenic factors tip the balance in favor of tumor angiogenesis. Among pro-angiogenic factors, vascular endothelial growth factor (VEGF) is a prominent target in therapeutic methods due to its strategic involvement in the formation of anomalous tumor vasculature. In addition, VEGF exhibits immune-regulatory properties which suppress immune cell antitumor activity. VEGF signaling through its receptors is an integral part of tumoral angiogenic approaches. A wide variety of medicines have been designed to target the ligands and receptors of this pro-angiogenic superfamily. Herein, we summarize the direct and indirect molecular mechanisms of VEGF to demonstrate its versatile role in the context of cancer angiogenesis and current transformative VEGF-targeted strategies interfering with tumor growth.
Collapse
Affiliation(s)
| | - Jale Yuzugulen
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus via Mersin 10, Turkey
| | | | | |
Collapse
|
29
|
Hasegawa H, Tanaka T, Kondo M, Teramoto K, Nakayama K, Hwang GW. Blood vessel remodeling in the cerebral cortex induced by binge alcohol intake in mice. Toxicol Res 2023; 39:169-177. [PMID: 36726835 PMCID: PMC9839917 DOI: 10.1007/s43188-022-00164-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Ethanol is toxic to the brain and causes various neurological disorders. Although ethanol can directly exert toxicity on neurons, it also acts on other cell types in the central nervous system. Blood vessel endothelial cells interact with, and are affected by blood ethanol. However, the effects of ethanol on the vascular structures of the brain have not been well documented. In this study, we examined the effects of binge levels of ethanol on brain vasculature. Immunostaining analysis indicated structural alterations of blood vessels in the cerebral cortex, which became more tortuous than those in the control mice after ethanol administration. The interaction between the blood vessels and astrocytes decreased, especially in the upper layers of the cerebral cortex. Messenger RNA expression analysis revealed a unique downregulation of Vegfa mRNA encoding vascular endothelial growth factor (VEGF)-A among VEGF, angiopoietin, endothelin family angiogenic and blood vessel remodeling factors. The expression of three proteoglycan core proteins, glypican-5, neurocan, and serglycin, was also altered after ethanol administration. Thus, binge levels of ethanol affect the expression of VEGF-A and blood vessel-supporting proteoglycans, resulting in changes in the vascular structure of the cerebral cortex. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-022-00164-y.
Collapse
Affiliation(s)
- Hiroshi Hasegawa
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Toshiya Tanaka
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Mari Kondo
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Koji Teramoto
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Kei Nakayama
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Gi-Wook Hwang
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 9818558 Japan
| |
Collapse
|
30
|
Sung DC, Chen M, Dominguez MH, Mahadevan A, Chen X, Yang J, Gao S, Ren AA, Tang AT, Mericko P, Patton R, Lee M, Jannaway M, Nottebaum A, Vestweber D, Scallan JP, Kahn ML. Sinusoidal and lymphatic vessel growth is controlled by reciprocal VEGF-C-CDH5 inhibition. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1006-1021. [PMID: 36910472 PMCID: PMC9997205 DOI: 10.1038/s44161-022-00147-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 09/14/2022] [Indexed: 11/12/2022]
Abstract
Sinusoids are specialized, low pressure blood vessels in the liver, bone marrow, and spleen required for definitive hematopoiesis. Unlike other blood endothelial cells (ECs), sinusoidal ECs express high levels of VEGFR3. VEGFR3 and its ligand VEGF-C are known to support lymphatic growth, but their function in sinusoidal vessels is unknown. In this study, we define a reciprocal VEGF-C/VEGFR3-CDH5 (VE-cadherin) signaling axis that controls growth of both sinusoidal and lymphatic vessels. Loss of VEGF-C or VEGFR3 resulted in cutaneous edema, reduced fetal liver size, and bloodless bone marrow due to impaired lymphatic and sinusoidal vessel growth. Mice with membrane-retained VE-cadherin conferred identical lymphatic and sinusoidal defects, suggesting that VE-cadherin opposes VEGF-C/VEGFR3 signaling. In developing mice, loss of VE-cadherin rescued defects in sinusoidal and lymphatic growth caused by loss of VEGFR3 but not loss of VEGF-C, findings explained by potentiated VEGF-C/VEGFR2 signaling in VEGFR3-deficient lymphatic ECs. Mechanistically, VEGF-C/VEGFR3 signaling induces VE-cadherin endocytosis and loss of function via SRC-mediated phosphorylation, while VE-cadherin prevents VEGFR3 endocytosis required for optimal receptor signaling. These findings establish an essential role for VEGF-C/VEGFR3 signaling during sinusoidal vascular growth, identify VE-cadherin as a powerful negative regulator of VEGF-C signaling that acts through both VEGFR3 and VEGFR2 receptors, and suggest that negative regulation of VE-cadherin is required for effective VEGF-C/VEGFR3 signaling during growth of sinusoidal and lymphatic vessels. Manipulation of this reciprocal negative regulatory mechanism, e.g. by reducing VE-cadherin function, may be used to stimulate therapeutic sinusoidal or lymphatic vessel growth.
Collapse
Affiliation(s)
- Derek C. Sung
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mei Chen
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Martin H. Dominguez
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Aparna Mahadevan
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaowen Chen
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jisheng Yang
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Siqi Gao
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Aileen A. Ren
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alan T. Tang
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patricia Mericko
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Raiyah Patton
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michelle Lee
- University Laboratory Animal Resources, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Melanie Jannaway
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Astrid Nottebaum
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | | | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Mark L. Kahn
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Althali NJ, Hentges KE. Genetic insights into non-syndromic Tetralogy of Fallot. Front Physiol 2022; 13:1012665. [PMID: 36277185 PMCID: PMC9582763 DOI: 10.3389/fphys.2022.1012665] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/13/2022] [Indexed: 10/17/2023] Open
Abstract
Congenital heart defects (CHD) include structural abnormalities of the heart or/and great vessels that are present at birth. CHD affects around 1% of all newborns worldwide. Tetralogy of Fallot (TOF) is the most prevalent cyanotic congenital cardiac abnormality, affecting three out of every 10,000 live infants with a prevalence rate of 5-10% of all congenital cardiac defects. The four hallmark characteristics of TOF are: right ventricular hypertrophy, pulmonary stenosis, ventricular septal defect, and overriding aorta. Approximately 20% of cases of TOF are associated with a known disease or chromosomal abnormality, with the remaining 80% of TOF cases being non-syndromic, with no known aetiology. Relatively few TOF patients have been studied, and little is known about critical causative genes for non-syndromic TOF. However, rare genetic variants have been identified as significant risk factors for CHD, and are likely to cause some cases of TOF. Therefore, this review aims to provide an update on well-characterized genes and the most recent variants identified for non-syndromic TOF.
Collapse
Affiliation(s)
- Nouf J. Althali
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Biology Department, Science College, King Khalid University, Abha, Saudi Arabia
| | - Kathryn E. Hentges
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
32
|
Hénon P, Kowalczyk M, Aries A, Vignon C, Trébuchet G, Lahlil R. Industrialized GMP Production of CD34 + Cells (ProtheraCytes®) at Clinical Scale for Treatment of Ischemic Cardiac Diseases Is Feasible and Safe. Stem Cell Rev Rep 2022; 18:1614-1626. [PMID: 35420389 PMCID: PMC9209364 DOI: 10.1007/s12015-022-10373-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2022] [Indexed: 02/08/2023]
Abstract
Regenerative medicine now needs to pass a crucial turning point, from academic research to the market. Several sources/types of cells have been experimented with, more or less successfully. CD34+ cells have demonstrated multipotent or even pluripotent capacities, making them good candidates for regenerative medicine, particularly for treating heart diseases. Strongly encouraged by the results we achieved in a pilot study using CD34+ stem cells in patients with poor-prognosis acute myocardial infarcts (AMIs), we soon began the development of an industrialized platform making use of a closed automated device (StemXpand®) and a disposable kit (StemPack®) for the large-scale expansion of CD34+ cells with reproducible good manufacturing practice (GMP). This scalable platform can produce expanded CD34+ cells (ProtheraCytes®) of sufficient quality that, interestingly, express early markers of the cardiac and endothelial pathways and early cardiac-mesoderm markers. They also contain CD34+ pluripotent cells characterized as very small embryonic-like stem cells (VSELs), capable of differentiating under appropriate stimuli into different tissue lineages, including endothelial and cardiomyocytic ones.
Collapse
Affiliation(s)
| | | | - Anne Aries
- Institut de Recherche en Hématologie et Transplantation, Hôpital du Hasenrain, 87 Avenue d'Altkirch, Mulhouse, France
| | | | | | - Rachid Lahlil
- Institut de Recherche en Hématologie et Transplantation, Hôpital du Hasenrain, 87 Avenue d'Altkirch, Mulhouse, France
| |
Collapse
|
33
|
Koh GY. Apelin drives maintenance and expansion of the vascular niche in intestinal crypts. NATURE CARDIOVASCULAR RESEARCH 2022; 1:410-412. [PMID: 39195944 DOI: 10.1038/s44161-022-00071-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Gou Young Koh
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
34
|
Abstract
Karaman et al. (2022. J. Exp. Med.https://doi.org/10.1084/jem.20210565) examined the differential effects of the conditional deletion of genes encoding each VEGF receptor, VEGFR1, VEGFR2 and VEGFR 3, as well as combinations thereof in mice. The results highlight the crosstalk between receptors in different organs and emphasize the importance of VEGF receptor expression and interplay in vascular heterogeneity.
Collapse
Affiliation(s)
- Pin Li
- Department of Pathology, University of California, San Diego, La Jolla, CA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Napoleone Ferrara
- Department of Pathology, University of California, San Diego, La Jolla, CA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|