1
|
Zhang Z, Zhao R, Wu X, Ma Y, He Y. Research progress on the correlation between corneal neovascularization and lymphangiogenesis (Review). Mol Med Rep 2025; 31:47. [PMID: 39635819 PMCID: PMC11638739 DOI: 10.3892/mmr.2024.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
The cornea is a clear connective tissue membrane at the front of the outer layer of the eyeball wall. It plays a crucial role in the refractive system of the eyeball, making it essential to maintain its transparency. Neovascularization and lymphangiogenesis in the cornea significantly impact corneal transparency and immune privilege. The growth of corneal neovascularization (CNV) and corneal lymphangiogenesis (CL) vessels is interconnected yet independent. Currently, there is a substantial amount of clinical and experimental research on CNV and CL vessels. However, due to the relatively recent focus on CL vessel research compared with CNV research, most scholars tend to concentrate on CNV, with few articles offering a comprehensive comparison and discussion of the two processes. The present review emphasizes the similarities and differences between CNV and CL and summarizes recent research progress on their correlation in animal models, growth characteristics, cytokine effects and related diseases.
Collapse
Affiliation(s)
- Zhaochen Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Rongxuan Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Xuhui Wu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yunkun Ma
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yuxi He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
2
|
Puy C, Moellmer SA, Pang J, Vu HH, Melrose AR, Lorentz CU, Tucker EI, Shatzel JJ, Keshari RS, Lupu F, Gailani D, McCarty OJT. Coagulation factor XI regulates endothelial cell permeability and barrier function in vitro and in vivo. Blood 2024; 144:1821-1833. [PMID: 39158072 DOI: 10.1182/blood.2023022257] [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: 08/18/2023] [Revised: 06/26/2024] [Accepted: 07/21/2024] [Indexed: 08/20/2024] Open
Abstract
ABSTRACT Loss of endothelial barrier function contributes to the pathophysiology of many inflammatory diseases. Coagulation factor XI (FXI) plays a regulatory role in inflammation. Although activation of FXI increases vascular permeability in vivo, the mechanism by which FXI or its activated form FXIa disrupts endothelial barrier function is unknown. We investigated the role of FXIa in human umbilical vein endothelial cell (HUVEC) or human aortic endothelial cell (HAEC) permeability. The expression patterns of vascular endothelial (VE)-cadherin and other proteins of interest were examined by western blot or immunofluorescence. Endothelial cell permeability was analyzed by Transwell assay. We demonstrate that FXIa increases endothelial cell permeability by inducing cleavage of the VE-cadherin extracellular domain, releasing a soluble fragment. The activation of a disintegrin and metalloproteinase 10 (ADAM10) mediates the FXIa-dependent cleavage of VE-cadherin, because adding an ADAM10 inhibitor prevented the cleavage of VE-cadherin induced by FXIa. The binding of FXIa with plasminogen activator inhibitor 1 and very low-density lipoprotein receptor on HUVEC or HAEC surfaces activates vascular endothelial growth receptor factor 2 (VEGFR2). The activation of VEGFR2 triggers the mitogen-activated protein kinase (MAPK) signaling pathway and promotes the expression of active ADAM10 on the cell surface. In a pilot experiment using an established baboon model of sepsis, the inhibition of FXI activation significantly decreased the levels of soluble VE-cadherin to preserve barrier function. This study reveals a novel pathway by which FXIa regulates vascular permeability. The effect of FXIa on barrier function may be another way by which FXIa contributes to the development of inflammatory diseases.
Collapse
Affiliation(s)
- Cristina Puy
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Samantha A Moellmer
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Helen H Vu
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Alexander R Melrose
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Christina U Lorentz
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
- Aronora, Inc, Portland, OR
| | - Erik I Tucker
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
- Aronora, Inc, Portland, OR
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR
| | - Ravi S Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - David Gailani
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR
| |
Collapse
|
3
|
Sun B, Li Q, Xiao X, Zhang J, Zhou Y, Huang Y, Gao J, Cao X. The loach haplotype-resolved genome and the identification of Mex3a involved in fish air breathing. CELL GENOMICS 2024; 4:100670. [PMID: 39389021 PMCID: PMC11602589 DOI: 10.1016/j.xgen.2024.100670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/30/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Fish air breathing is crucial for the transition of vertebrates from water to land. So far, the genes involved in fish air breathing have not been well identified. Here, we performed gene enrichment analysis of positively selected genes (PSGs) in loach (Misgurnus anguillicaudatus, an air-breathing fish) in comparison to Triplophysa tibetana (a non-air-breathing fish), haplotype-resolved genome assembly of the loach, and gene evolutionary analysis of air-breathing and non-air-breathing fishes and found that the PSG mex3a originated from ancient air-breathing fish species. Deletion of Mex3a impaired loach air-breathing capacity by inhibiting angiogenesis through its interaction with T-box transcription factor 20. Mex3a overexpression significantly promoted angiogenesis. Structural analysis and point mutation revealed the critical role of the 201st amino acid in loach Mex3a for angiogenesis. Our findings innovatively indicate that the ancient mex3a is a fish air-breathing gene, which holds significance for understanding fish air breathing and provides a valuable resource for cultivating hypoxia-tolerant fish varieties.
Collapse
Affiliation(s)
- Bing Sun
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingshan Li
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Xinxin Xiao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianwei Zhang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Ying Zhou
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuwei Huang
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian Gao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China.
| | - Xiaojuan Cao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
4
|
Shen J, Su X, Wang S, Wang Z, Zhong C, Huang Y, Duan S. RhoJ: an emerging biomarker and target in cancer research and treatment. Cancer Gene Ther 2024; 31:1454-1464. [PMID: 38858534 DOI: 10.1038/s41417-024-00792-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024]
Abstract
RhoJ is a Rho GTPase that belongs to the Cdc42 subfamily and has a molecular weight of approximately 21 kDa. It can activate the p21-activated kinase family either directly or indirectly, influencing the activity of various downstream effectors and playing a role in regulating the cytoskeleton, cell movement, and cell cycle. RhoJ's expression and activity are controlled by multiple upstream factors at different levels, including expression, subcellular localization, and activation. High RhoJ expression is generally associated with a poor prognosis for cancer patients and is mainly due to an increased number of tumor blood vessels and abnormal expression in malignant cells. RhoJ promotes tumor progression through several pathways, particularly in tumor angiogenesis and drug resistance. Clinical data also indicates that high RhoJ expression is closely linked to the pathological features of tumor malignancy. There are various cancer treatment methods that target RhoJ signaling, such as direct binding to inhibit the RhoJ effector pocket, inhibiting RhoJ expression, blocking RhoJ upstream and downstream signals, and indirectly inhibiting RhoJ's effect. RhoJ is an emerging cancer biomarker and a significant target for future cancer clinical research and drug development.
Collapse
Affiliation(s)
- Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Xinming Su
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shana Wang
- Department of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zehua Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Chenming Zhong
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Yi Huang
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Wei W, Jiang T, Hu F, Liu H. Tibial transverse transport combined with platelet-rich plasma sustained-release microspheres activates the VEGFA/VEGFR2 pathway to promote microcirculatory reconstruction in diabetic foot ulcer. Growth Factors 2024; 42:128-144. [PMID: 39329304 DOI: 10.1080/08977194.2024.2407318] [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: 03/26/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
This study proposes to investigate the therapeutic efficacy and mechanism of combining tibial transverse transport (TTT) with platelet-rich plasma (PRP) for diabetic foot ulcer (DFU). The diabetic rabbit model was constructed with Streptozotocin, which was intervened with TTT and PRP. PRP injection combined with TTT significantly promoted vascularisation and enhanced CD31, VEGFA, and VEGFR2 expressions compared to traditional TTT. However, the VEGFR2 inhibitor suppressed these phenomena. In the in vitro injury model, PRP reversed the diminished human umbilical vein endothelial cells (HUVECs) function and vascularisation caused by high-glucose damage. Additionally, PRP reduced inflammation and oxidative stress (approximately 47% ROS level) and enhanced VEGFA and VEGFR2 expression in HUVECs. However, the knockdown of VEGFR2 reversed the effect of PRP. In conclusion, TTT combined with intraosseous flap injection of PRP sustained-release microspheres activated the VEGFA/VEGFR2 pathway to promote microcirculatory reconstruction in DFU. These findings may provide new potential therapeutic strategies for DFU.
Collapse
Affiliation(s)
- Weiqiang Wei
- Department of Orthopaedics, The Fourth Hospital of Changsha, Changsha, China
| | - Tenglong Jiang
- Department of Orthopaedics, The Fourth Hospital of Changsha, Changsha, China
| | - Fan Hu
- Department of Orthopaedics, The Fourth Hospital of Changsha, Changsha, China
| | - Hong Liu
- Department of Orthopaedics, The Fourth Hospital of Changsha, Changsha, China
| |
Collapse
|
6
|
Al-Ostoot FH, Salah S, Khanum SA. An Overview of Cancer Biology, Pathophysiological Development and It's Treatment Modalities: Current Challenges of Cancer anti-Angiogenic Therapy. Cancer Invest 2024; 42:559-604. [PMID: 38874308 DOI: 10.1080/07357907.2024.2361295] [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: 02/17/2021] [Revised: 11/22/2021] [Accepted: 05/25/2024] [Indexed: 06/15/2024]
Abstract
A number of conditions and factors can cause the transformation of normal cells in the body into malignant tissue by changing the normal functions of a wide range of regulatory, apoptotic, and signal transduction pathways. Despite the current deficiency in fully understanding the mechanism of cancer action accurately and clearly, numerous genes and proteins that are causally involved in the initiation, progression, and metastasis of cancer have been identified. But due to the lack of space and the abundance of details on this complex topic, we have emphasized here more recent advances in our understanding of the principles implied tumor cell transformation, development, invasion, angiogenesis, and metastasis. Inhibition of angiogenesis is a significant strategy for the treatment of various solid tumors, that essentially depend on cutting or at least limiting the supply of blood to micro-regions of tumors, leading to pan-hypoxia and pan-necrosis inside solid tumor tissues. Researchers have continued to enhance the efficiency of anti-angiogenic drugs over the past two decades, to identify their potential in the drug interaction, and to discover reasonable interpretations for possible resistance to treatment. In this review, we have discussed an overview of cancer history and recent methods use in cancer therapy, focusing on anti-angiogenic inhibitors targeting angiogenesis formation. Further, this review has explained the molecular mechanism of action of these anti-angiogenic inhibitors in various tumor types and their limitations use. In addition, we described the synergistic mechanisms of immunotherapy and anti-angiogenic therapy and summarizes current clinical trials of these combinations. Many phase III trials found that combining immunotherapy and anti-angiogenic therapy improved survival. Therefore, targeting the source supply of cancer cells to grow and spread with new anti-angiogenic agents in combination with different conventional therapy is a novel method to reduce cancer progression. The aim of this paper is to overview the varying concepts of cancer focusing on mechanisms involved in tumor angiogenesis and provide an overview of the recent trends in anti-angiogenic strategies for cancer therapy.
Collapse
Affiliation(s)
- Fares Hezam Al-Ostoot
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India
- Department of Biochemistry, Faculty of Education & Science, Albaydha University, Al-Baydha, Yemen
| | - Salma Salah
- Faculty of Medicine and Health Sciences, Thamar University, Dhamar, Yemen
| | - Shaukath Ara Khanum
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India
| |
Collapse
|
7
|
Nan W, He Y, Shen S, Wu M, Wang S, Zhang Y. BMP4 inhibits corneal neovascularization by interfering with tip cells in angiogenesis. Exp Eye Res 2023; 237:109680. [PMID: 37858608 DOI: 10.1016/j.exer.2023.109680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Corneal neovascularization (CNV) can lead to impaired corneal transparency, resulting in vision loss or blindness. The primary pathological mechanism underlying CNV is an imbalance between pro-angiogenic and anti-angiogenic factors, with inflammation playing a crucial role. Notably, a vascular endothelial growth factor(VEGF)-A gradient triggers the selection of single endothelial cells(ECs) into primary tip cells that guide sprouting, while a dynamic balance between tip and stalk cells maintains a specific ratio to promote CNV. Despite the central importance of tip-stalk cell selection and shuffling, the underlying mechanisms remain poorly understood. In this study, we examined the effects of bone morphogenetic protein 4 (BMP4) on VEGF-A-induced lumen formation in human umbilical vein endothelial cells (HUVECs) and CD34-stained tip cell formation. In vivo, BMP4 inhibited CNV caused by corneal sutures. This process was achieved by BMP4 decreasing the protein expression of VEGF-A and VEGFR2 in corneal tissue after corneal suture injury. By observing the ultrastructure of the cornea, BMP4 inhibited the sprouting of tip cells and brought forward the appearance of intussusception. Meanwhile, BMP4 attenuated the inflammatory response by inhibiting neutrophil extracellular traps (NETs)formation through the NADPH oxidase-2(NOX-2)pathway. Our results indicate that BMP4 inhibits the formation of tip cells by reducing the generation of NETs, disrupting the dynamic balance of tip and stalk cells and thereby inhibiting CNV, suggesting that BMP4 may be a potential therapeutic target for CNV.
Collapse
Affiliation(s)
- Weijin Nan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Yuxi He
- Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Sitong Shen
- Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Meiliang Wu
- Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Shurong Wang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, 130000, China
| | - Yan Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
8
|
Li J, Huang H, Xu S, Fan M, Wang K, Wang X, Zhang J, Huang S, Gatt A, Liu J. Complement factor H inhibits endothelial cell migration through suppression of STAT3 signaling. Exp Ther Med 2023; 26:408. [PMID: 37522066 PMCID: PMC10375431 DOI: 10.3892/etm.2023.12107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 06/13/2023] [Indexed: 08/01/2023] Open
Abstract
Complement factor H (CFH), a major soluble inhibitor of complement, is a plasma protein that directly interacts with the endothelium of blood vessels. Mutations in the CFH gene lead to diseases associated with excessive angiogenesis; however, the underlying mechanisms are unknown. The present study aimed to determine the effects of CFH on endothelial cells and to explore the underlying mechanisms. The adenoviral plasmid expressing CFH was transduced into HepG2 cells, and the culture medium supernatant was collected and co-cultured with human umbilical vein endothelial cells (HUVECs). Cell proliferation was measured by CCK8 and MTT assays, and cell migration was measured by wound healing and Transwell assays. Reverse transcription-quantitative PCR was performed to detect gene transcription. Western blotting was used to determine protein levels. The results revealed that CFH can inhibit migration, but not viability, of HUVECs. In addition, CFH did not significantly alter MAPK or TGF-β signaling, whereas it decreased STAT3 phosphorylation in HUVECs. Furthermore, CFH failed to reduce migration of HUVECs, with inhibition of STAT3 signaling by STATTIC or activation of STAT3 signaling by overexpression of STAT3 (Y705D) compromising CFH-inhibited HUVEC migration. CFH also decreased the expression levels of vascular endothelial growth factor receptor 2, a downstream effector of STAT3 mediating endothelial cell migration. In conclusion, the present study suggested that CFH may be a potential therapeutic target for angiogenesis-related diseases.
Collapse
Affiliation(s)
- Jiang Li
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Hong Huang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Shanhu Xu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Mengge Fan
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Kaili Wang
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Xia Wang
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Jiao Zhang
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Shengshi Huang
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Alex Gatt
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta
- Haematology Laboratory, Department of Pathology, Mater Dei Hospital, Msida, MSD 2080, Malta
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
9
|
Middelkoop MA, Don EE, Hehenkamp WJK, Polman NJ, Griffioen AW, Huirne JAF. Angiogenesis in abnormal uterine bleeding: a narrative review. Hum Reprod Update 2023; 29:457-485. [PMID: 36857162 PMCID: PMC10320491 DOI: 10.1093/humupd/dmad004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/12/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Abnormal uterine bleeding (AUB) has a significant socioeconomic impact since it considerably impacts quality of life. Therapeutic options are frequently based on trial and error and do not target disease aetiology. Pathophysiological insight in this disease is required for the development of novel treatment options. If no underlying cause is found for the AUB (e.g. fibroids, adenomyosis, polyps), endometrial-AUB (AUB-E) is usually caused by a primary endometrium disorder. When AUB is induced by prescribed (exogenous) hormones, it is classified as iatrogenic-AUB (AUB-I). Considering vascular modulation and function, AUB-E and AUB-I both could potentially result from abnormal vascularization in the endometrium due to alterations in the process of angiogenesis and vascular maturation. OBJECTIVE AND RATIONALE We aim to investigate the fundamental role of angiogenesis and vascular maturation in patients with AUB and hypothesize that aberrant endometrial angiogenesis has an important role in the aetiology of both AUB-E and AUB-I, possibly through different mechanisms. SEARCH METHODS A systematic literature search was performed until September 2021 in the Cochrane Library Databases, Embase, PubMed, and Web of Science, with search terms such as angiogenesis and abnormal uterine bleeding. Included studies reported on angiogenesis in the endometrium of premenopausal women with AUB-E or AUB-I. Case reports, letters, reviews, editorial articles, and studies on AUB with causes classified by the International Federation of Gynecology and Obstetrics as myometrial, oncological, or infectious, were excluded. Study quality was assessed by risk of bias, using the Cochrane tool and the Newcastle-Ottawa Scale. OUTCOMES Thirty-five out of 2158 articles were included. In patients with AUB-E, vascular endothelial growth factor A and its receptors (1 and 2), as well as the angiopoietin-1:angiopoietin-2 ratio and Tie-1, were significantly increased. Several studies reported on the differential expression of other pro- and antiangiogenic factors in patients with AUB-E, suggesting aberrant vascular maturation and impaired vessel integrity. Overall, endometrial microvessel density (MVD) was comparable in patients with AUB-E and controls. Interestingly, patients with AUB-I showed a higher MVD and higher expression of proangiogenic factors when compared to controls, in particular after short-term hormone exposure. This effect was gradually lost after longer-term exposure, while alterations in vessel maturation were observed after both short- and long-term exposures. WIDER IMPLICATIONS AUB-E and AUB-I are most likely associated with aberrant endometrial angiogenesis and impaired vessel maturation. This review supports existing evidence that increased proangiogenic and decreased antiangiogenic factors cause impaired vessel maturation, resulting in more fragile and permeable vessels. This matches our hypothesis and these mechanisms appear to play an important role in the pathophysiology of AUB-E and AUB-I. Exploring the alterations in angiogenesis in these patients could provide treatment targets for AUB.
Collapse
Affiliation(s)
- Mei-An Middelkoop
- Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
| | - Emma E Don
- Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
| | - Wouter J K Hehenkamp
- Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
| | - Nicole J Polman
- Department of Obstetrics and Gynaecology, Flevoziekenhuis, Almere, the Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Judith A F Huirne
- Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Urbiola-Salvador V, Lima de Souza S, Grešner P, Qureshi T, Chen Z. Plasma Proteomics Unveil Novel Immune Signatures and Biomarkers upon SARS-CoV-2 Infection. Int J Mol Sci 2023; 24:ijms24076276. [PMID: 37047248 PMCID: PMC10093853 DOI: 10.3390/ijms24076276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/07/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Several elements have an impact on COVID-19, including comorbidities, age and sex. To determine the protein profile changes in peripheral blood caused by a SARS-CoV-2 infection, a proximity extension assay was used to quantify 1387 proteins in plasma samples among 28 Finnish patients with COVID-19 with and without comorbidities and their controls. Key immune signatures, including CD4 and CD28, were changed in patients with comorbidities. Importantly, several unreported elevated proteins in patients with COVID-19, such as RBP2 and BST2, which show anti-microbial activity, along with proteins involved in extracellular matrix remodeling, including MATN2 and COL6A3, were identified. RNF41 was downregulated in patients compared to healthy controls. Our study demonstrates that SARS-CoV-2 infection causes distinct plasma protein changes in the presence of comorbidities despite the interpatient heterogeneity, and several novel potential biomarkers associated with a SARS-CoV-2 infection alone and in the presence of comorbidities were identified. Protein changes linked to the generation of SARS-CoV-2-specific antibodies, long-term effects and potential association with post-COVID-19 condition were revealed. Further study to characterize the identified plasma protein changes from larger cohorts with more diverse ethnicities of patients with COVID-19 combined with functional studies will facilitate the identification of novel diagnostic, prognostic biomarkers and potential therapeutic targets for patients with COVID-19.
Collapse
Affiliation(s)
- Víctor Urbiola-Salvador
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, 80-307 Gdańsk, Pomerania, Poland
| | - Suiane Lima de Souza
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, North Ostrobothnia, Finland
| | - Peter Grešner
- Department of Translational Oncology, Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, Medical University of Gdańsk, 80-211 Gdańsk, Pomerania, Poland
| | - Talha Qureshi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, North Ostrobothnia, Finland
| | - Zhi Chen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, North Ostrobothnia, Finland
- Correspondence:
| |
Collapse
|
11
|
BMP4 aggravates mitochondrial dysfunction of HRMECs. Heliyon 2023; 9:e13824. [PMID: 36895361 PMCID: PMC9988459 DOI: 10.1016/j.heliyon.2023.e13824] [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: 09/20/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Mitochondria are important places for the oxidative phosphorylation of glucose and the maintenance of cell oxidation and antioxidant stability. However, mitochondrial dysfunction causes cell dysfunction. Meanwhile, retinal vascular endothelial cell dysfunction may cause vascular inflammation, hemorrhage, angiogenesis, and other manifestations. Our previous studies have shown that Bone morphogenetic protein 4 (BMP4) is an important target for the treatment of retinal neovascularization, but the mechanism remains unclear. Therefore, our study aims to observe the effects of BMP4 on vascular endothelial cells and hopes to provide a new target for diabetic retinopathy. 4-Hydroxynonenal (4HNE), a kind of lipid peroxide, was used to induce the oxidative stress model. Human retinal microvascular endothelial cells (HRMECs) were randomly divided into control, 4HNE, negative control, and siBMP4 groups. Si-BMP4 significantly reduced leukocyte adhesion and 4HNE-induced high ROS level and restored the mitochondrial membrane potential and OCR. This indicates that BMP4 plays an important role in inducing leukocyte adhesion, oxidative stress, and mitochondrial dysfunction. The relationship between BMP4 and retinal vascular endothelial cell dysfunction is preliminarily confirmed by this study. Mitochondrial dysfunction and oxidative stress may be involved in BMP4-mediated retinal vascular endothelial cell dysfunction.
Collapse
|
12
|
Gnanaguru G, Tabor SJ, Bonilla GM, Sadreyev R, Yuda K, Köhl J, Connor KM. Microglia refine developing retinal astrocytic and vascular networks through the complement C3/C3aR axis. Development 2023; 150:dev201047. [PMID: 36762625 PMCID: PMC10110418 DOI: 10.1242/dev.201047] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
Microglia, a resident immune cell of the central nervous system (CNS), play a pivotal role in facilitating neurovascular development through mechanisms that are not fully understood. Previous reports indicate a role for microglia in regulating astrocyte density. This current work resolves the mechanism through which microglia facilitate astrocyte spatial patterning and superficial vascular bed formation in the neuroretina during development. Ablation of microglia increased astrocyte density and altered spatial patterning. Mechanistically, we show that microglia regulate the formation of the spatially organized astrocyte template required for subsequent vascular growth, through the complement C3/C3aR axis during neuroretinal development. Lack of C3 or C3aR hindered the developmental phagocytic removal of astrocyte bodies and resulted in increased astrocyte density. In addition, increased astrocyte density was associated with elevated proangiogenic extracellular matrix gene expression in C3- and C3aR-deficient retinas, resulting in increased vascular density. These data demonstrate that microglia regulate developmental astrocyte and vascular network spatial patterning in the neuroretina via the complement axis.
Collapse
Affiliation(s)
- Gopalan Gnanaguru
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Steven J. Tabor
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Gracia M. Bonilla
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Kentaro Yuda
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kip M. Connor
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
13
|
Vliora M, Ravelli C, Grillo E, Corsini M, Flouris AD, Mitola S. The impact of adipokines on vascular networks in adipose tissue. Cytokine Growth Factor Rev 2023; 69:61-72. [PMID: 35953434 DOI: 10.1016/j.cytogfr.2022.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023]
Abstract
Adipose tissue (AT) is a highly active and plastic endocrine organ. It secretes numerous soluble molecules known as adipokines, which act locally to AT control the remodel and homeostasis or exert pleiotropic functions in different peripheral organs. Aberrant production or loss of certain adipokines contributes to AT dysfunction associated with metabolic disorders, including obesity. The AT plasticity is strictly related to tissue vascularization. Angiogenesis supports the AT expansion, while regression of blood vessels is associated with AT hypoxia, which in turn mediates tissue inflammation, fibrosis and metabolic dysfunction. Several adipokines can regulate endothelial cell functions and are endowed with either pro- or anti-angiogenic properties. Here we address the role of adipokines in the regulation of angiogenesis. A better understanding of the link between adipokines and angiogenesis will open the way for novel therapeutic approaches to treat obesity and metabolic diseases.
Collapse
Affiliation(s)
- Maria Vliora
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece; Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Andreas D Flouris
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy.
| |
Collapse
|
14
|
Grillo E, Ravelli C, Colleluori G, D'Agostino F, Domenichini M, Giordano A, Mitola S. Role of gremlin-1 in the pathophysiology of the adipose tissues. Cytokine Growth Factor Rev 2023; 69:51-60. [PMID: 36155165 DOI: 10.1016/j.cytogfr.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 02/07/2023]
Abstract
Gremlin-1 is a secreted bone morphogenetic protein (BMP) antagonist playing a pivotal role in the regulation of tissue formation and embryonic development. Since its first identification in 1997, gremlin-1 has been shown to be a multifunctional factor involved in wound healing, inflammation, cancer and tissue fibrosis. Among others, the activity of gremlin-1 is mediated by its interaction with BMPs or with membrane receptors such as the vascular endothelial growth factor receptor 2 (VEGFR2) or heparan sulfate proteoglycans (HSPGs). Growing evidence has highlighted a central role of gremlin-1 in the homeostasis of the adipose tissue (AT). Of note, gremlin-1 is involved in AT dysfunction during type 2 diabetes, obesity and non-alcoholic fatty liver disease (NAFLD) metabolic disorders. In this review we discuss recent findings on gremlin-1 involvement in AT biology, with particular attention to its role in metabolic diseases, to highlight its potential as a prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Georgia Colleluori
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Francesco D'Agostino
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mattia Domenichini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
15
|
Sano T, Nakajima T, Senda KA, Nakano S, Yamato M, Ikeda Y, Zeng H, Kawabe JI, Matsunaga YT. Image-based crosstalk analysis of cell-cell interactions during sprouting angiogenesis using blood-vessel-on-a-chip. Stem Cell Res Ther 2022; 13:532. [PMID: 36575469 PMCID: PMC9795717 DOI: 10.1186/s13287-022-03223-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Sprouting angiogenesis is an important mechanism for morphogenetic phenomena, including organ development, wound healing, and tissue regeneration. In regenerative medicine, therapeutic angiogenesis is a clinical solution for recovery from ischemic diseases. Mesenchymal stem cells (MSCs) have been clinically used given their pro-angiogenic effects. MSCs are reported to promote angiogenesis by differentiating into pericytes or other vascular cells or through cell-cell communication using multiple protein-protein interactions. However, how MSCs physically contact and move around ECs to keep the sprouting angiogenesis active remains unknown. METHODS We proposed a novel framework of EC-MSC crosstalk analysis using human umbilical vein endothelial cells (HUVECs) and MSCs obtained from mice subcutaneous adipose tissue on a 3D in vitro model, microvessel-on-a-chip, which allows cell-to-tissue level study. The microvessels were fabricated and cultured for 10 days in a collagen matrix where MSCs were embedded. RESULTS Immunofluorescence imaging using a confocal laser microscope showed that MSCs smoothed the surface of the microvessel and elongated the angiogenic sprouts by binding to the microvessel's specific microstructures. Additionally, three-dimensional modeling of HUVEC-MSC intersections revealed that MSCs were selectively located around protrusions or roots of angiogenic sprouts, whose surface curvature was excessively low or high, respectively. CONCLUSIONS The combination of our microvessel-on-a-chip system for 3D co-culture and image-based crosstalk analysis demonstrated that MSCs are selectively localized to concave-convex surfaces on scaffold structures and that they are responsible for the activation and stabilization of capillary vessels.
Collapse
Affiliation(s)
- Takanori Sano
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Tadaaki Nakajima
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan ,grid.268441.d0000 0001 1033 6139Department of Science, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027 Japan
| | - Koharu Alicia Senda
- Hiroo Gakuen Junior and Senior High School, 5-1-14 Minami Azabu, Minato-ku, Tokyo, 106-0047 Japan
| | - Shizuka Nakano
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Mizuho Yamato
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Yukinori Ikeda
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Hedele Zeng
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Jun-ichi Kawabe
- grid.252427.40000 0000 8638 2724Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-higashi, Asahikawa, Hokkaido 078-8510 Japan
| | - Yukiko T. Matsunaga
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| |
Collapse
|
16
|
de Oliveira NK, Ferraz EP, Rosin FCP, Correa L, Deboni MCZ. Poly-ε-caprolactone/poly(rotaxane) seeded with human dental pulp stem cells or osteoblasts promotes angiogenesis: a chorioallantoic membrane assay. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2051-2066. [PMID: 35719115 DOI: 10.1080/09205063.2022.2091372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 06/15/2023]
Abstract
Biomaterials used for tissue regeneration should ideally provide a favorable environment for cell proliferation and differentiation. Angiogenesis is crucial for supplying oxygen and nutrients necessary for cellular survival at implantation sites. The aim of this study was to evaluate the overall angiogenesis response of a poly ε-caprolactone/poly (rotaxane) blend (poly-blend) carried by human dental pulp stem cells (hDPSCs) or osteoblasts (OB) seeded in the chorioallantoic membranes (CAM) of fertilized chicken eggs on embryonic day 7. They were classified into the following intervention groups: (a) poly(polymeric blend disks free of cells); (b) hDPSC seeded onto CAM; (c) poly/hDPSC (where hDPSCs were seeded onto poly-blend); (d) poly/OB (where osteoblasts were seeded onto poly); (e) OB (where hDPSCs differentiated into osteoblasts were seeded onto CAM); and (f) a negative control when a sterilized silicone ring free of cells or polymer was inserted into CAM. On embryonic day 14, the quantitative and qualitative characteristics of the blood vessels in the CAMs were analyzed macroscopically and microscopically. Macroscopic examination showed that the Poly/hDPSC samples exhibited an increased medium vessel density. Additionally, microscopic observations showed that the Poly/hDPSC group and poly alone resulted in a large lumen area of vascularization. Thus, poly ε-caprolactone/poly (rotaxane) did not impair angiogenesis. Furthermore, poly-blend carried by stem cells of dental pulp origin shows a better vasculogenic potential, which is essential for regenerative therapies.
Collapse
Affiliation(s)
| | - Emanuela Prado Ferraz
- Oral Surgery Department, School of Dentistry, University of São Paulo - FOUSP, São Paulo, SP, Brazil
| | | | - Luciana Correa
- Pathology Department, School of Dentistry, University of São Paulo - FOUSP, São Paulo, SP, Brazil
| | | |
Collapse
|
17
|
Wu E, Fan X, Tang T, Li J, Wang J, Liu X, Zungar Z, Ren J, Wu C, Shen B. Biomarkers discovery for endometrial cancer: A graph convolutional sample network method. Comput Biol Med 2022; 150:106200. [PMID: 37859290 DOI: 10.1016/j.compbiomed.2022.106200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/20/2022] [Accepted: 10/09/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Endometrial carcinoma is the sixth most common cancer in women worldwide. Importantly, endometrial cancer is among the few types of cancers with patient mortality that is still increasing, which indicates that the improvement in its diagnosis and treatment is still urgent. Moreover, biomarker discovery is essential for precise classification and prognostic prediction of endometrial cancer. METHODS A novel graph convolutional sample network method was used to identify and validate biomarkers for the classification of endometrial cancer. The sample networks were first constructed for each sample, and the gene pairs with high frequencies were identified to construct a subtype-specific network. Putative biomarkers were then screened using the highest degrees in the subtype-specific network. Finally, simplified sample networks are constructed using the biomarkers for the graph convolutional network (GCN) training and prediction. RESULTS Putative biomarkers (23) were identified using the novel bioinformatics model. These biomarkers were then rationalised with functional analyses and were found to be correlated to disease survival with network entropy characterisation. These biomarkers will be helpful in future investigations of the molecular mechanisms and therapeutic targets of endometrial cancers. CONCLUSIONS A novel bioinformatics model combining sample network construction with GCN modelling is proposed and validated for biomarker discovery in endometrial cancer. The model can be generalized and applied to biomarker discovery in other complex diseases.
Collapse
Affiliation(s)
- Erman Wu
- Institutes for Systems Genetics, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xuemeng Fan
- Institutes for Systems Genetics, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Tong Tang
- Institutes for Systems Genetics, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China; Department of Computer Science and Information Technologies, Elviña Campus, University of A Coruña, A Coruña, Spain
| | - Jingjing Li
- Institutes for Systems Genetics, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Wang
- Institutes for Systems Genetics, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xingyun Liu
- Institutes for Systems Genetics, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zayatta Zungar
- School of Medicine, University of New England, Armidale, NSW, 2351, Australia
| | - Jiaojiao Ren
- School of Electronic Information and Electrical Engineering, Chengdu University, Chengdu, China
| | - Cong Wu
- Institutes for Systems Genetics, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Ibrutinib Inhibits Angiogenesis and Tumorigenesis in a BTK-Independent Manner. Pharmaceutics 2022; 14:pharmaceutics14091876. [PMID: 36145624 PMCID: PMC9506105 DOI: 10.3390/pharmaceutics14091876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/31/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
BTK inhibitor (BTKi) Ibrutinib carries an increased bleeding risk compared to more selective BTKis Acalabrutinib and Zanubrutinib, however, its impact on vascular endothelium remains unknown. In this study, we found that Ibrutinib induced stronger cytotoxic effect on endothelial cells than Zanubrutinib, however, Acalabrutinib cytotoxicity was extremely weak. RNA-seq, followed by KEGG analysis and quantitative RT-PCR validation, was conducted to identify the differential apoptotic target genes of BTKis, leading to their distinct cytotoxic effects on endothelial cells, which showed that Ibrutinib and Zanubrutinib dramatically modulated the expression of critical apoptotic genes, GADD45B, FOS, and BCL2A1, among which FOS and GADD45B were upregulated more significantly by Ibrutinib than Zanubrutinib, however, Acalabrutinib downregulated BCL2A1 moderately and was not able to modulate the expression of FOS and GADD45B. Next, we performed in vitro angiogenesis assays and found that Ibrutinib was more able to induce endothelial dysfunction than Zanubrutinib via stimulating more BMP4 expression, however, Acalabrutinib had no such effect. Especially, the capacity of Ibrutinib to induce endothelial dysfunction can be antagonized by targeting BMP4. Accordingly, Ibrutinib, as an angiogenesis inhibitor, inhibited ovarian and breast cancer progression in vivo. Collectively, our findings addressed a novel molecular basis underlying Ibrutinib-induced endothelial cell dysfunction and suggested the potential application of Ibrutinib to treat angiogenesis-dependent cancers.
Collapse
|
19
|
Kutikhin AG, Shishkova DK, Velikanova EA, Sinitsky MY, Sinitskaya AV, Markova VE. Endothelial Dysfunction in the Context of Blood–Brain Barrier Modeling. J EVOL BIOCHEM PHYS+ 2022; 58:781-806. [PMID: 35789679 PMCID: PMC9243926 DOI: 10.1134/s0022093022030139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 01/04/2023]
Abstract
Here, we discuss pathophysiological approaches to the defining
of endothelial dysfunction criteria (i.e., endothelial activation,
impaired endothelial mechanotransduction, endothelial-to-mesenchymal
transition, reduced nitric oxide release, compromised endothelial
integrity, and loss of anti-thrombogenic properties) in different
in vitro and in vivo models. The canonical definition of endothelial
dysfunction includes insufficient production of vasodilators, pro-thrombotic
and pro-inflammatory activation of endothelial cells, and pathologically
increased endothelial permeability. Among the clinical consequences
of endothelial dysfunction are arterial hypertension, macro- and
microangiopathy, and microalbuminuria. We propose to extend the definition
of endothelial dysfunction by adding altered endothelial mechanotransduction
and endothelial-to-mesenchymal transition to its criteria. Albeit
interleukin-6, interleukin-8, and MCP-1/CCL2 dictate the pathogenic
paracrine effects of dysfunctional endothelial cells and are therefore
reliable endothelial dysfunction biomarkers in vitro, they are non-specific
for endothelial cells and cannot be used for the diagnostics of
endothelial dysfunction in vivo. Conceptual improvements in the
existing methods to model endothelial dysfunction, specifically,
in relation to the blood–brain barrier, include endothelial cell
culturing under pulsatile flow, collagen IV coating of flow chambers,
and endothelial lysate collection from the blood vessels of laboratory
animals in situ for the subsequent gene and protein expression profiling.
Combined with the simulation of paracrine effects by using conditioned
medium from dysfunctional endothelial cells, these flow-sensitive
models have a high physiological relevance, bringing the experimental
conditions to the physiological scenario.
Collapse
Affiliation(s)
- A. G. Kutikhin
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - D. K. Shishkova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - E. A. Velikanova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - M. Yu. Sinitsky
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A. V. Sinitskaya
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - V. E. Markova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
20
|
Ozisik O, Ehrhart F, Evelo CT, Mantovani A, Baudot A. Overlap of vitamin A and vitamin D target genes with CAKUT-related processes. F1000Res 2022; 10:395. [PMID: 35528959 PMCID: PMC9051587 DOI: 10.12688/f1000research.51018.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
Congenital Anomalies of the Kidney and Urinary Tract (CAKUT) are a group of abnormalities affecting the kidneys and their outflow tracts. CAKUT patients display a large clinical variability as well as a complex aetiology. Only 5% to 20% of the cases have a monogenic origin. It is thereby suspected that interactions of both genetic and environmental factors contribute to the disease. Vitamins are among the environmental factors that are considered for CAKUT aetiology. In this study, we aimed to investigate whether vitamin A or vitamin D could have a role in CAKUT aetiology. For this purpose we collected vitamin A and vitamin D target genes and computed their overlap with CAKUT-related gene sets. We observed limited overlap between vitamin D targets and CAKUT-related gene sets. We however observed that vitamin A target genes significantly overlap with multiple CAKUT-related gene sets, including CAKUT causal and differentially expressed genes, and genes involved in renal system development. Overall, these results indicate that an excess or deficiency of vitamin A might be relevant to a broad range of urogenital abnormalities.
Collapse
Affiliation(s)
- Ozan Ozisik
- Aix Marseille University, Inserm, MMG, Marseille, 13385, France
| | - Friederike Ehrhart
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, 6200 MD, The Netherlands
- Department of Bioinformatics, NUTRIM/MHeNs, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Chris T. Evelo
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | | | - Anaïs Baudot
- Aix Marseille University, Inserm, MMG, Marseille, 13385, France
- Barcelona Supercomputing Center (BSC), Barcelona, 08034, Spain
| |
Collapse
|
21
|
Miller B, Sewell-Loftin MK. Mechanoregulation of Vascular Endothelial Growth Factor Receptor 2 in Angiogenesis. Front Cardiovasc Med 2022; 8:804934. [PMID: 35087885 PMCID: PMC8787114 DOI: 10.3389/fcvm.2021.804934] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
The endothelial cells that compose the vascular system in the body display a wide range of mechanotransductive behaviors and responses to biomechanical stimuli, which act in concert to control overall blood vessel structure and function. Such mechanosensitive activities allow blood vessels to constrict, dilate, grow, or remodel as needed during development as well as normal physiological functions, and the same processes can be dysregulated in various disease states. Mechanotransduction represents cellular responses to mechanical forces, translating such factors into chemical or electrical signals which alter the activation of various cell signaling pathways. Understanding how biomechanical forces drive vascular growth in healthy and diseased tissues could create new therapeutic strategies that would either enhance or halt these processes to assist with treatments of different diseases. In the cardiovascular system, new blood vessel formation from preexisting vasculature, in a process known as angiogenesis, is driven by vascular endothelial growth factor (VEGF) binding to VEGF receptor 2 (VEGFR-2) which promotes blood vessel development. However, physical forces such as shear stress, matrix stiffness, and interstitial flow are also major drivers and effectors of angiogenesis, and new research suggests that mechanical forces may regulate VEGFR-2 phosphorylation. In fact, VEGFR-2 activation has been linked to known mechanobiological agents including ERK/MAPK, c-Src, Rho/ROCK, and YAP/TAZ. In vascular disease states, endothelial cells can be subjected to altered mechanical stimuli which affect the pathways that control angiogenesis. Both normalizing and arresting angiogenesis associated with tumor growth have been strategies for anti-cancer treatments. In the field of regenerative medicine, harnessing biomechanical regulation of angiogenesis could enhance vascularization strategies for treating a variety of cardiovascular diseases, including ischemia or permit development of novel tissue engineering scaffolds. This review will focus on the impact of VEGFR-2 mechanosignaling in endothelial cells (ECs) and its interaction with other mechanotransductive pathways, as well as presenting a discussion on the relationship between VEGFR-2 activation and biomechanical forces in the extracellular matrix (ECM) that can help treat diseases with dysfunctional vascular growth.
Collapse
Affiliation(s)
- Bronte Miller
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
22
|
Patient-specific iPSC-derived endothelial cells reveal aberrant p38 MAPK signaling in atypical hemolytic uremic syndrome. Stem Cell Reports 2021; 16:2305-2319. [PMID: 34388364 PMCID: PMC8452517 DOI: 10.1016/j.stemcr.2021.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 11/24/2022] Open
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a rare disease associated with high morbidity and mortality. Existing evidence suggests that the central pathogenesis to aHUS might be endothelial cell damage. Nevertheless, the role of endothelial cell alterations in aHUS has not been well characterized and the underlying mechanisms remain unclear. Utilizing an induced pluripotent stem cell-derived endothelial cell (iPSC-EC) model, we showed that anti-complement factor H autoantibody-associated aHUS patient-specific iPSC-ECs exhibited an intrinsic defect in endothelial functions. Stimulation using aHUS serums exacerbated endothelial dysfunctions, leading to cell apoptosis in iPSC-ECs. Importantly, we identified p38 as a novel signaling pathway contributing to endothelial dysfunctions in aHUS. These results illustrate that iPSC-ECs can be a reliable model to recapitulate EC pathological features, thus providing a unique platform for gaining mechanistic insights into EC injury in aHUS. Our findings highlight that the p38 MAPK signaling pathway can be a therapeutic target for treatment of aHUS. aHUS patient-specific iPSC-ECs exhibit intrinsic defect in endothelial functions Stimulation using aHUS serums exacerbates EC dysfunctions and causes EC apoptosis p38 signaling contributes to EC dysfunctions in anti-CFH Ab-associated aHUS
Collapse
|
23
|
Bartonella henselae Persistence within Mesenchymal Stromal Cells Enhances Endothelial Cell Activation and Infectibility That Amplifies the Angiogenic Process. Infect Immun 2021; 89:e0014121. [PMID: 34031126 DOI: 10.1128/iai.00141-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Some bacterial pathogens can manipulate the angiogenic response, suppressing or inducing it for their own ends. In humans, Bartonella henselae is associated with cat-scratch disease and vasculoproliferative disorders such as bacillary angiomatosis and bacillary peliosis. Although endothelial cells (ECs) support the pathogenesis of B. henselae, the mechanisms by which B. henselae induces EC activation are not completely clear, as well as the possible contributions of other cells recruited at the site of infection. Mesenchymal stromal cells (MSCs) are endowed with angiogenic potential and play a dual role in infections, exerting antimicrobial properties but also acting as a shelter for pathogens. Here, we delved into the role of MSCs as a reservoir of B. henselae and modulator of EC functions. B. henselae readily infected MSCs and survived in perinuclearly bound vacuoles for up to 8 days. Infection enhanced MSC proliferation and the expression of epidermal growth factor receptor (EGFR), Toll-like receptor 2 (TLR2), and nucleotide-binding oligomerization domain-containing protein 1 (NOD1), proteins that are involved in bacterial internalization and cytokine production. Secretome analysis revealed that infected MSCs secreted higher levels of the proangiogenic factors vascular endothelial growth factor (VEGF), fibroblast growth factor 7 (FGF-7), matrix metallopeptidase 9 (MMP-9), placental growth factor (PIGF), serpin E1, thrombospondin 1 (TSP-1), urokinase-type plasminogen activator (uPA), interleukin 6 (IL-6), platelet-derived growth factor D (PDGF-D), chemokine ligand 5 (CCL5), and C-X-C motif chemokine ligand 8 (CXCL8). Supernatants from B. henselae-infected MSCs increased the susceptibility of ECs to B. henselae infection and enhanced EC proliferation, invasion, and reorganization in tube-like structures. Altogether, these results indicate MSCs as a still underestimated niche for persistent B. henselae infection and reveal MSC-EC cross talk that may contribute to exacerbate bacterium-induced angiogenesis and granuloma formation.
Collapse
|
24
|
The BMP Pathway in Blood Vessel and Lymphatic Vessel Biology. Int J Mol Sci 2021; 22:ijms22126364. [PMID: 34198654 PMCID: PMC8232321 DOI: 10.3390/ijms22126364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) were originally identified as the active components in bone extracts that can induce ectopic bone formation. In recent decades, their key role has broadly expanded beyond bone physiology and pathology. Nowadays, the BMP pathway is considered an important player in vascular signaling. Indeed, mutations in genes encoding different components of the BMP pathway cause various severe vascular diseases. Their signaling contributes to the morphological, functional and molecular heterogeneity among endothelial cells in different vessel types such as arteries, veins, lymphatic vessels and capillaries within different organs. The BMP pathway is a remarkably fine-tuned pathway. As a result, its signaling output in the vessel wall critically depends on the cellular context, which includes flow hemodynamics, interplay with other vascular signaling cascades and the interaction of endothelial cells with peri-endothelial cells and the surrounding matrix. In this review, the emerging role of BMP signaling in lymphatic vessel biology will be highlighted within the framework of BMP signaling in the circulatory vasculature.
Collapse
|
25
|
Ozisik O, Ehrhart F, Evelo CT, Mantovani A, Baudot A. Overlap of vitamin A and vitamin D target genes with CAKUT-related processes. F1000Res 2021; 10:395. [PMID: 35528959 PMCID: PMC9051587 DOI: 10.12688/f1000research.51018.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2022] [Indexed: 08/24/2023] Open
Abstract
Congenital Anomalies of the Kidney and Urinary Tract (CAKUT) are a group of abnormalities affecting the kidneys and their outflow tracts. CAKUT patients display a large clinical variability as well as a complex aetiology. Only 5% to 20% of the cases have a monogenic origin. It is thereby suspected that interactions of both genetic and environmental factors contribute to the disease. Vitamins are among the environmental factors that are considered for CAKUT aetiology. In this study, we aimed to investigate whether vitamin A or vitamin D could have a role in CAKUT aetiology. For this purpose we collected vitamin A and vitamin D target genes and computed their overlap with CAKUT-related gene sets. We observed limited overlap between vitamin D targets and CAKUT-related gene sets. We however observed that vitamin A target genes significantly overlap with multiple CAKUT-related gene sets, including CAKUT causal and differentially expressed genes, and genes involved in renal system development. Overall, these results indicate that an excess or deficiency of vitamin A might be relevant to a broad range of urogenital abnormalities.
Collapse
Affiliation(s)
- Ozan Ozisik
- Aix Marseille University, Inserm, MMG, Marseille, 13385, France
| | - Friederike Ehrhart
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, 6200 MD, The Netherlands
- Department of Bioinformatics, NUTRIM/MHeNs, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Chris T. Evelo
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | | | - Anaïs Baudot
- Aix Marseille University, Inserm, MMG, Marseille, 13385, France
- Barcelona Supercomputing Center (BSC), Barcelona, 08034, Spain
| |
Collapse
|
26
|
Yang D, Yang X, Dai F, Wang Y, Yang Y, Hu M, Cheng Y. The Role of Bone Morphogenetic Protein 4 in Ovarian Function and Diseases. Reprod Sci 2021; 28:3316-3330. [PMID: 33966186 DOI: 10.1007/s43032-021-00600-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/22/2021] [Indexed: 12/19/2022]
Abstract
Bone morphogenetic proteins (BMPs) are the largest subfamily of the transforming growth factor-β (TGF-β) superfamily. BMP4 is a secreted protein that was originally identified due to its role in bone and cartilage development. Over the past decades, extensive literature has indicated that BMP4 and its receptors are widely expressed in the ovary. Dysregulation of BMP4 expression may play a vital role in follicular development, polycystic ovary syndrome (PCOS), and ovarian cancer. In this review, we summarized the expression pattern of BMP4 in the ovary, focused on the role of BMP4 in follicular development and steroidogenesis, and discussed the role of BMP4 in ovarian diseases such as polycystic ovary syndrome and ovarian cancer. Some studies have shown that the expression of BMP4 in the ovary is spatiotemporal and species specific, but the effects of BMP4 seem to be similar in follicular development of different species. In addition, BMP4 is involved in the development of hyperandrogenemia in PCOS and drug resistance in ovarian cancer, but further research is still needed to clarify the specific mechanisms.
Collapse
Affiliation(s)
- Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiao Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yanqing Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Yang
- School of Physics & Technology, Key Laboratory of Artificial Micro/Nano Structure of Ministry of Education, Wuhan University, Wuhan, 430072, China.
| | - Min Hu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
27
|
Hancock PC, Koduru SV, Sun M, Ravnic DJ. Induction of scaffold angiogenesis by recipient vasculature precision micropuncture. Microvasc Res 2021; 134:104121. [PMID: 33309646 DOI: 10.1016/j.mvr.2020.104121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/12/2020] [Accepted: 12/08/2020] [Indexed: 12/01/2022]
Abstract
The success of engineered tissues continues to be limited by time to vascularization and perfusion. Here, we studied the effects of precision injury to a recipient macrovasculature in promoting neovessel formation in an adjacently placed scaffold. Segmental 60 μm diameter micropunctures (MP) were created in the recipient rat femoral artery and vein followed by coverage with a simple collagen scaffold. Scaffolds were harvested at 24, 48, 72, and 96 h post-implantation for detailed analysis. Those placed on top of an MP segment showed an earlier and more robust cellular infiltration, including both endothelial cells (CD31) and macrophages (F4/80), compared to internal non-micropunctured control limbs (p < 0.05). At the 96-hour timepoint, MP scaffolds demonstrated an increase in physiologic perfusion (p < 0.003) and a 2.5-fold increase in capillary network formation (p < 0.001). These were attributed to an overall upsurge in small vessel quantity. Furthermore, MP positioned scaffolds demonstrated significant increases in many modulators of angiogenesis, including VEGFR2 and Tie-2 despite a decrease in HIF-1α at all timepoints. This study highlights a novel microsurgical approach that can be used to rapidly vascularize or inosculate contiguously placed scaffolds and grafts. Thereby, offering an easily translatable route towards the creation of thicker and more clinically relevant engineered tissues.
Collapse
Affiliation(s)
- Patrick C Hancock
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Srinivas V Koduru
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA; Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Mingjie Sun
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA; Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Dino J Ravnic
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA; Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
28
|
VEGF-Independent Activation of Müller Cells by the Vitreous from Proliferative Diabetic Retinopathy Patients. Int J Mol Sci 2021; 22:ijms22042179. [PMID: 33671690 PMCID: PMC7926720 DOI: 10.3390/ijms22042179] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
Proliferative diabetic retinopathy (PDR), a major complication of diabetes mellitus, results from an inflammation-sustained interplay among endothelial cells, neurons, and glia. Even though anti-vascular endothelial growth factor (VEGF) interventions represent the therapeutic option for PDR, they are only partially efficacious. In PDR, Müller cells undergo reactive gliosis, produce inflammatory cytokines/chemokines, and contribute to scar formation and retinal neovascularization. However, the impact of anti-VEGF interventions on Müller cell activation has not been fully elucidated. Here, we show that treatment of MIO-M1 Müller cells with vitreous obtained from PDR patients stimulates cell proliferation and motility, and activates various intracellular signaling pathways. This leads to cytokine/chemokine upregulation, a response that was not mimicked by treatment with recombinant VEGF nor inhibited by the anti-VEGF drug ranibizumab. In contrast, fibroblast growth factor-2 (FGF2) induced a significant overexpression of various cytokines/chemokines in MIO-M1 cells. In addition, the FGF receptor tyrosine kinase inhibitor BGJ398, the pan-FGF trap NSC12, the heparin-binding protein antagonist N-tert-butyloxycarbonyl-Phe-Leu-Phe-Leu-Phe Boc2, and the anti-inflammatory hydrocortisone all inhibited Müller cell activation mediated by PDR vitreous. These findings point to a role for various modulators beside VEGF in Müller cell activation and pave the way to the search for novel therapeutic strategies in PDR.
Collapse
|
29
|
Corsini M, Ravelli C, Grillo E, Dell'Era P, Presta M, Mitola S. Simultaneously characterization of tumoral angiogenesis and vasculogenesis in stem cell-derived teratomas. Exp Cell Res 2021; 400:112490. [PMID: 33484747 DOI: 10.1016/j.yexcr.2021.112490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/02/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022]
Abstract
Tumor neovascularization may occur via both angiogenic and vasculogenic events. In order to investigate the vessel formation during tumor growth, we developed a novel experimental model that takes into account the differentiative and tumorigenic properties of Embryonic Stem cells (ESCs). Leukemia Inhibitory Factor-deprived murine ESCs were grafted on the top of the chick embryo chorionallantoic membrane (CAM) in ovo. Cell grafts progressively grew, forming a vascularized mass within 10 days. At this stage, the grafts are formed by cells with differentiative features representative of all three germ layers, thus originating teratomas, a germinal cell tumor. In addition, ESC supports neovascular events by recruiting host capillaries from surrounding tissue that infiltrates the tumor mass. Moreover, immunofluorescence studies demonstrate that perfused active blood vessels within the tumor are of both avian and murine origin because of the simultaneous occurrence of angiogenic and vasculogenic events. In conclusion, the chick embryo ESC/CAM-derived teratoma model may represent a useful approach to investigate both vasculogenic and angiogenic events during tumor growth and for the study of natural and synthetic modulators of the two processes.
Collapse
Affiliation(s)
- Michela Corsini
- Department of Molecular and Translational Medicine, Via Branze 39, 25123, Brescia, University of Brescia, Italy; Laboratory for Preventive and Personalized Medicine (MPP Lab), University of Brescia, Italy.
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, Via Branze 39, 25123, Brescia, University of Brescia, Italy; Laboratory for Preventive and Personalized Medicine (MPP Lab), University of Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, Via Branze 39, 25123, Brescia, University of Brescia, Italy
| | - Patrizia Dell'Era
- Department of Molecular and Translational Medicine, Via Branze 39, 25123, Brescia, University of Brescia, Italy; cFRU Lab, Università degli Studi di Brescia, Viale Europa 11, 25123, Brescia, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, Via Branze 39, 25123, Brescia, University of Brescia, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, Via Branze 39, 25123, Brescia, University of Brescia, Italy; Laboratory for Preventive and Personalized Medicine (MPP Lab), University of Brescia, Italy.
| |
Collapse
|
30
|
Grillo E, Corsini M, Ravelli C, di Somma M, Zammataro L, Monti E, Presta M, Mitola S. A novel variant of VEGFR2 identified by a pan-cancer screening of recurrent somatic mutations in the catalytic domain of tyrosine kinase receptors enhances tumor growth and metastasis. Cancer Lett 2020; 496:84-92. [PMID: 33035615 DOI: 10.1016/j.canlet.2020.09.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 01/10/2023]
Abstract
In cancer genomics, recurrence of mutations in gene families that share homologous domains has recently emerged as a reliable indicator of functional impact and can be exploited to reveal the pro-oncogenic effect of previously uncharacterized variants. Pan-cancer analyses of mutation hotspots in the catalytic domain of a subset of tyrosine kinase receptors revealed that two infrequent mutations of VEGFR2 (R1051Q and D1052N) recur in analogous proteins and correlate with reduced patient survival. Functional validation showed that both R1051Q and D1052N mutations increase the enzymatic activity of VEGFR2. The expression of VEGFR2R1051Q potentiates the PI3K/Akt signaling axis in cancer cells, increasing their tumorigenic potential in vitro and in vivo. In addition, it confers to cancer cells an increased sensitivity to the VEGFR2-targeted tyrosine kinase inhibitor Linifanib. In the context of an efficacious application of anti-cancer targeted therapies, these findings indicate that the screening for uncharacterized mutations, like VEGFR2R1051Q, may help to predict patient prognosis and drug response, with significant clinical implications.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy.
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy; Laboratory for Preventive and Personalized Medicine (MPP Lab), University of Brescia, 25123, Italy
| | - Margherita di Somma
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Luca Zammataro
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| | - Eugenio Monti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy; Laboratory for Preventive and Personalized Medicine (MPP Lab), University of Brescia, 25123, Italy.
| |
Collapse
|
31
|
Marín-Ramos NI, Thein TZ, Ghaghada KB, Chen TC, Giannotta SL, Hofman FM. miR-18a Inhibits BMP4 and HIF-1α Normalizing Brain Arteriovenous Malformations. Circ Res 2020; 127:e210-e231. [PMID: 32755283 DOI: 10.1161/circresaha.119.316317] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Brain arteriovenous malformations (AVMs) are abnormal tangles of vessels where arteries and veins directly connect without intervening capillary nets, increasing the risk of intracerebral hemorrhage and stroke. Current treatments are highly invasive and often not feasible. Thus, effective noninvasive treatments are needed. We previously showed that AVM-brain endothelial cells (BECs) secreted higher VEGF (vascular endothelial growth factor) and lower TSP-1 (thrombospondin-1) levels than control BEC; and that microRNA-18a (miR-18a) normalized AVM-BEC function and phenotype, although its mechanism remained unclear. OBJECTIVE To elucidate the mechanism of action and potential clinical application of miR-18a as an effective noninvasive treatment to selectively restore the phenotype and functionality of AVM vasculature. METHODS AND RESULTS The molecular pathways affected by miR-18a in patient-derived BECs and AVM-BECs were determined by Western blot, RT-qPCR (quantitative reverse transcription polymerase chain reaction), ELISA, co-IP, immunostaining, knockdown and overexpression studies, flow cytometry, and luciferase reporter assays. miR-18a was shown to increase TSP-1 and decrease VEGF by reducing PAI-1 (plasminogen activator inhibitor-1/SERPINE1) levels. Furthermore, miR-18a decreased the expression of BMP4 (bone morphogenetic protein 4) and HIF-1α (hypoxia-inducible factor 1α), blocking the BMP4/ALK (activin-like kinase) 2/ALK1/ALK5 and Notch signaling pathways. As determined by Boyden chamber assays, miR-18a also reduced the abnormal AVM-BEC invasiveness, which correlated with a decrease in MMP2 (matrix metalloproteinase 2), MMP9, and ADAM10 (ADAM metallopeptidase domain 10) levels. In vivo pharmacokinetic studies showed that miR-18a reaches the brain following intravenous and intranasal administration. Intranasal co-delivery of miR-18a and NEO100, a good manufacturing practices-quality form of perillyl alcohol, improved the pharmacokinetic profile of miR-18a in the brain without affecting its pharmacological properties. Ultra-high-resolution computed tomography angiography and immunostaining studies in an Mgp-/- AVM mouse model showed that miR-18a decreased abnormal cerebral vasculature and restored the functionality of the bone marrow, lungs, spleen, and liver. CONCLUSIONS miR-18a may have significant clinical value in preventing, reducing, and potentially reversing AVM.
Collapse
Affiliation(s)
- Nagore I Marín-Ramos
- Departments of Neurosurgery (N.I.M.-R., T.Z.T., T.C.C., S.L.G.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Thu Zan Thein
- Departments of Neurosurgery (N.I.M.-R., T.Z.T., T.C.C., S.L.G.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Ketan B Ghaghada
- Department of Pediatric Radiology, Texas Children's Hospital, Houston (K.B.G.)
| | - Thomas C Chen
- Departments of Neurosurgery (N.I.M.-R., T.Z.T., T.C.C., S.L.G.), Keck School of Medicine, University of Southern California, Los Angeles.,Departments of Pathology (T.C.C., F.M.H.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Steven L Giannotta
- Departments of Neurosurgery (N.I.M.-R., T.Z.T., T.C.C., S.L.G.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Florence M Hofman
- Departments of Pathology (T.C.C., F.M.H.), Keck School of Medicine, University of Southern California, Los Angeles
| |
Collapse
|
32
|
Zhang S, Yue J, Ge Z, Xie Y, Zhang M, Jiang L. Activation of CXCR7 alleviates cardiac insufficiency after myocardial infarction by promoting angiogenesis and reducing apoptosis. Biomed Pharmacother 2020; 127:110168. [PMID: 32361166 DOI: 10.1016/j.biopha.2020.110168] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/04/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is an important pathway for revascularization of ischemic tissues after acute myocardial infarction (AMI). It is unclear what role CXCR7 plays in angiogenesis in the ischemic area after AMI, although some researchers have shown that the activation of CXCR7 protectsthe heart under those conditions. Here, we hypothesize that the activation of CXCR7 promotes angiogenesis, reduces cell apoptosis and alleviates cardiac deficiency after AMI. C57BL/6 J mice were subjected to AMI and treated with TC14012 (10 mg/kg) for 24 days. HUVECs were cultured in a hypoxic (2% O2) environment to generate a model of hypoxia. CXCR7 was knocked down in HUVECs by sh-CXCR7 transfection, and CXCR7 was activated by TC14012 (30 μM) treatment. The results showed that CXCR7 was downregulated in infarcted heart tissue and hypoxic HUVECs. The global activation of CXCR7 may alleviate the decrease in cardiac function indexes - (ejection fraction and fraction shortening), and reduce infarct size after AMI.. Moreover, CXCR7 activation has been shown to enhance the level of angiogenesis in ischemic heart tissue. In vitro, hypoxia-induced angiogenic functional loss and apoptosis are aggravated by CXCR7 knockdown in HUVECs. Both angiogenic impairment and cell apoptosis are rescued by CXCR7 activation. In conclusion, the present study indicates that activation of CXCR7 plays an important protective role for ischemic cells in hypoxic endothelial cells and AMI model mice by promoting angiogenesis and reducing apoptosis, which suggests that CXCR7 may be a potential therapeutic target to rescue the ischemic myocardium..
Collapse
Affiliation(s)
- Sheng Zhang
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, China
| | - Jingwen Yue
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, China
| | - Zhuowang Ge
- Division of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, China
| | - Yi Xie
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, China
| | - Min Zhang
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, China.
| | - Li Jiang
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, China.
| |
Collapse
|
33
|
Li Y, Liu Z, Tang Y, Feng W, Zhao C, Liao J, Zhang C, Chen H, Ren Y, Dong S, Liu Y, Hu N, Huang W. Schnurri-3 regulates BMP9-induced osteogenic differentiation and angiogenesis of human amniotic mesenchymal stem cells through Runx2 and VEGF. Cell Death Dis 2020; 11:72. [PMID: 31996667 PMCID: PMC6989499 DOI: 10.1038/s41419-020-2279-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
Human amniotic mesenchymal stem cells (hAMSCs) are multiple potent progenitor cells (MPCs) that can differentiate into different lineages (osteogenic, chondrogenic, and adipogenic cells) and have a favorable capacity for angiogenesis. Schnurri-3 (Shn3) is a large zinc finger protein related to Drosophila Shn, which is a critical mediator of postnatal bone formation. Bone morphogenetic protein 9 (BMP9), one of the most potent osteogenic BMPs, can strongly upregulate various osteogenesis- and angiogenesis-related factors in MSCs. It remains unclear how Shn3 is involved in BMP9-induced osteogenic differentiation coupled with angiogenesis in hAMSCs. In this investigation, we conducted a comprehensive study to identify the effect of Shn3 on BMP9-induced osteogenic differentiation and angiogenesis in hAMSCs and analyze the responsible signaling pathway. The results from in vitro and in vivo experimentation show that Shn3 notably inhibits BMP9-induced early and late osteogenic differentiation of hAMSCs, expression of osteogenesis-related factors, and subcutaneous ectopic bone formation from hAMSCs in nude mice. Shn3 also inhibited BMP9-induced angiogenic differentiation, expression of angiogenesis-related factors, and subcutaneous vascular invasion in mice. Mechanistically, we found that Shn3 prominently inhibited the expression of BMP9 and activation of the BMP/Smad and BMP/MAPK signaling pathways. In addition, we further found activity on runt-related transcription factor 2 (Runx2), vascular endothelial growth factor (VEGF), and the target genes shared by BMP and Shn3 signaling pathways. Silencing Shn3 could dramatically enhance the expression of Runx2, which directly regulates the downstream target VEGF to couple osteogenic differentiation with angiogenesis. To summarize, our findings suggested that Shn3 significantly inhibited the BMP9-induced osteogenic differentiation and angiogenesis in hAMSCs. The effect of Shn3 was primarily seen through inhibition of the BMP/Smad signaling pathway and depressed expression of Runx2, which directly regulates VEGF, which couples BMP9-induced osteogenic differentiation with angiogenesis.
Collapse
Affiliation(s)
- Yuwan Li
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ziming Liu
- Institute of Sports Medicine of China, Peking University Third Hospital, Beijing, 100191, China
| | - Yaping Tang
- Department of Stomatology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Wei Feng
- Laboratory of Skeletal Development and Regeneration, School of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Chen Zhao
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Junyi Liao
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chengmin Zhang
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Hong Chen
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Youliang Ren
- Department of Orthopaedics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Shiwu Dong
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yi Liu
- Department of Orthopaedics, the First Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Ning Hu
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Huang
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
34
|
Marshall KM, Kanczler JM, Oreffo ROC. Evolving applications of the egg: chorioallantoic membrane assay and ex vivo organotypic culture of materials for bone tissue engineering. J Tissue Eng 2020; 11:2041731420942734. [PMID: 33194169 PMCID: PMC7594486 DOI: 10.1177/2041731420942734] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/26/2020] [Indexed: 01/03/2023] Open
Abstract
The chick chorioallantoic membrane model has been around for over a century, applied in angiogenic, oncology, dental and xenograft research. Despite its often perceived archaic, redolent history, the chorioallantoic membrane assay offers new and exciting opportunities for material and growth factor evaluation in bone tissue engineering. Currently, superior/improved experimental methodology for the chorioallantoic membrane assay are difficult to identify, given an absence of scientific consensus in defining experimental approaches, including timing of inoculation with materials and the analysis of results. In addition, critically, regulatory and welfare issues impact upon experimental designs. Given such disparate points, this review details recent research using the ex vivo chorioallantoic membrane assay and the ex vivo organotypic culture to advance the field of bone tissue engineering, and highlights potential areas of improvement for their application based on recent developments within our group and the tissue engineering field.
Collapse
Affiliation(s)
- Karen M Marshall
- Bone and Joint Research Group, Centre for Human
Development, Stem Cells and Regeneration, Institute of Developmental Sciences,
University of Southampton, Southampton, UK
| | - Janos M Kanczler
- Bone and Joint Research Group, Centre for Human
Development, Stem Cells and Regeneration, Institute of Developmental Sciences,
University of Southampton, Southampton, UK
| | - Richard OC Oreffo
- Bone and Joint Research Group, Centre for Human
Development, Stem Cells and Regeneration, Institute of Developmental Sciences,
University of Southampton, Southampton, UK
| |
Collapse
|
35
|
Di Somma M, Schaafsma W, Grillo E, Vliora M, Dakou E, Corsini M, Ravelli C, Ronca R, Sakellariou P, Vanparijs J, Castro B, Mitola S. Natural Histogel-Based Bio-Scaffolds for Sustaining Angiogenesis in Beige Adipose Tissue. Cells 2019; 8:cells8111457. [PMID: 31752157 PMCID: PMC6912328 DOI: 10.3390/cells8111457] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 12/17/2022] Open
Abstract
In the treatment of obesity and its related disorders, one of the measures adopted is weight reduction by controlling nutrition and increasing physical activity. A valid alternative to restore the physiological function of the human body could be the increase of energy consumption by inducing the browning of adipose tissue. To this purpose, we tested the ability of Histogel, a natural mixture of glycosaminoglycans isolated from animal Wharton jelly, to sustain the differentiation of adipose derived mesenchymal cells (ADSCs) into brown-like cells expressing UCP-1. Differentiated cells show a higher energy metabolism compared to undifferentiated mesenchymal cells. Furthermore, Histogel acts as a pro-angiogenic matrix, induces endothelial cell proliferation and sprouting in a three-dimensional gel in vitro, and stimulates neovascularization when applied in vivo on top of the chicken embryo chorioallantoic membrane or injected subcutaneously in mice. In addition to the pro-angiogenic activity of Histogel, also the ADSC derived beige cells contribute to activating endothelial cells. These data led us to propose Histogel as a promising scaffold for the modulation of the thermogenic behavior of adipose tissue. Indeed, Histogel simultaneously supports the acquisition of brown tissue markers and activates the vasculature process necessary for the correct function of the thermogenic tissue. Thus, Histogel represents a valid candidate for the development of bioscaffolds to increase the amount of brown adipose tissue in patients with metabolic disorders.
Collapse
Affiliation(s)
- Margherita Di Somma
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Wandert Schaafsma
- Histocell, S.L.Parque Tecnológico 801A, 2o 48160 Derio—BIZKAIA, Spain; (W.S.); (B.C.)
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Maria Vliora
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
- FAME Laboratory, Department of Exercise Science, University of Thessaly, 38221 Trikala, Greece;
| | - Eleni Dakou
- Laboratory of Cell Genetics, Department of Biology, Faculty of Science and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium;
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Paraskevi Sakellariou
- FAME Laboratory, Department of Exercise Science, University of Thessaly, 38221 Trikala, Greece;
| | - Jef Vanparijs
- Department of Human Physiology, Faculty of Physical Education and Physical Therapy, Vrije Universiteit Brussel, 1050 Brussels, Belgium;
| | - Begona Castro
- Histocell, S.L.Parque Tecnológico 801A, 2o 48160 Derio—BIZKAIA, Spain; (W.S.); (B.C.)
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
- Correspondence:
| |
Collapse
|