1
|
Algandaby MM, Esmat A, Nasrullah MZ, Alhakamy NA, Abdel-Naim AB, Rashad OM, Elhady SS, Eltamany EE. LC-MS based metabolic profiling and wound healing activity of a chitosan nanoparticle-loaded formula of Teucrium polium in diabetic rats. Biomed Pharmacother 2023; 168:115626. [PMID: 37852098 DOI: 10.1016/j.biopha.2023.115626] [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: 08/05/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/20/2023] Open
Abstract
Healing of wounds is the most deteriorating diabetic experience. Felty germander (Teucrium polium) possesses antioxidant, anti-inflammatory and antimicrobial activities that could accelerate wound healing. Further, nanohydrogels help quicken healing and are ideal biomaterials for drug delivery. In the current study, the chemical profiling, and standardization of T. polium methanolic extract by LC-ESI/TOF/MS/MS and quantitative HPLC-DAD analyses were achieved. The wound healing enhancement in diabetic rats by T. polium nanopreparation (TP-NP) as chitosan nanogel (CS-NG) and investigating the potential mechanisms were investigated. The prepared hydrogel-based TP-NP were characterized with respect to particle size, zeta potential, pH, viscosity, and release of major components. LC-ESI/TOF/MS/MS metabolomic profiling of T. polium revealed the richness of the plant with phenolic compounds, particularly flavonoids. In addition, several terpenoids were detected. Kaempferol content of T. polium was estimated to be 7.85 ± 0.022 mg/ g of dry extract. The wound healing activity of TP-NP was explored in streptozotocin-induced diabetic rats. Diabetic animals were subjected to surgical wounding (1 cm diameter). Then they were divided in 5 groups (10 each). These included Group 1 (untreated control rats), Group 2 received the vehicle of CS-NG; Group 3 (0.5 g of TP prepared in hydrogel), Group 4 (0.5 g of TP-NP), Group 5 represented a positive control treated with 0.5 g of a commercial product. All treatments were applied topically for 21 days. Application of TP-NP on skin wounds of diabetic animals accelerated the healing process as evidenced by epithelium regeneration, formation of granulation tissue followed by epidermal proliferation, along with keratinization as verified by H&E. This was confirmed through enhanced collagen synthesis, as shown by raised hydroxyproline content and Col1A1 gene expression. Moreover, TP-NP significantly alleviated wound oxidative burst and diminished the expressions of inflammatory biomarkers. Meanwhile, TP-NP could enhance the expressions of transforming growth factor beta1 (TGF-β1), in addition to the angiogenic markers; vascular endothelia growth factor A (VEGFA) and platelet-derived growth factor receptor alpha (PDGFRα). Collectively, chitosan nanogel of T. polium accelerates wound healing in diabetic rats, which could be explained - at least partly - through alleviating oxidative stress and inflammation coupled with pro-angiogenic capabilities.
Collapse
Affiliation(s)
- Mardi M Algandaby
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ahmed Esmat
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Mohammed Z Nasrullah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ashraf B Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Omar M Rashad
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt.
| | - Sameh S Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Enas E Eltamany
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
2
|
Liu X, Chen J, Liu G, Zhang B, Jin X, Wang Y. MicroRNA-17-5p, a novel endothelial cell modulator, controls vascular re-endothelialization and neointimal lesion formation. Vascular 2023; 31:392-401. [PMID: 34958294 DOI: 10.1177/17085381211067672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND The functions of miR-17-5p in tumorigenesis have been explored. However, their functionalities in arterial endothelial cells (ECs) have not been investigated. Besides, the issue of vascular remodelling is barely addressed. OBJECTIVES The study aimed to determine the effect of overexpression or inhibition of miR-17-5p on arterial endothelial cells' (ECs) function and vascular remodelling in vitro and the rat carotid arteries model. METHODS Quantitative RT-PCR analysis was performed to examine the expression of miR-17-5p. Then, gain-of-function and loss-of-function approaches were employed to investigate the functional roles of miR-17-5p in cultured human coronary artery endothelial cells (HCAECs); further, TargetScan software analysis and luciferase reporter activity assay were performed to investigate the potential mechanism. Lastly, the results of the cell segment were verified in a rat carotid artery balloon injury model by Western blot analysis, measurement of the vascular cGMP level and plasma 8-iso-prostaglandin F2 (8-iso-PGF2) testing. Moreover, morphometric analysis was implemented to detect the re-endothelialization and neointimal formation in rat carotid artery after balloon injury. RESULTS This study firstly found that miR-17-5p expression was upregulated in the injured vascular walls and highly expressive in ECs; overexpression of miR-17-5p inhibited HCAECs' proliferation and migration, whereas miR-17-5p knockdown strengthened its proliferative and migratory roles, influenced inflammatory response, through regulating VEGRA and VEGFR2. It was found that miR-17-5p bind to VEGFA and VEGFR2 at the 3'UTR. Next, downregulation of miR-17-5p promotes re-endothelialization, and attenuates neointimal formation as measured by the I/M ratio (0.63±0.05 vs 1.45±0.06, antagomiR-17-5p vs. Lenti-NC, p < 0.05). In addition, the functional recovery of the endothelium was also accelerated by miR-17-5p knockdown. CONCLUSION Our study suggests that miR-17-5p is a feasible strategy for the selective modulation of endothelialization and vascular remodelling through regulating VEGFA and VEGFR2.
Collapse
Affiliation(s)
- Xiaopei Liu
- Department of Cardiology, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Jing Chen
- Department of Cardiology, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Gen Liu
- Department of Cardiology, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Bofang Zhang
- Department of Cardiology, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Xing Jin
- Department of Cardiology, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Yun Wang
- Department of Cardiology, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, 117921Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Narvaez Del Pilar O, Gacha Garay MJ, Chen J. Three-axis classification of mouse lung mesenchymal cells reveals two populations of myofibroblasts. Development 2022; 149:274755. [PMID: 35302583 PMCID: PMC8977099 DOI: 10.1242/dev.200081] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/21/2022] [Indexed: 10/18/2022]
Abstract
The mesenchyme consists of heterogeneous cell populations that support neighboring structures and are integral to intercellular signaling, but are poorly defined morphologically and molecularly. Leveraging single-cell RNA-sequencing, 3D imaging and lineage tracing, we classify the mouse lung mesenchyme into three proximal-distal axes that are associated with the endothelium, epithelium and interstitium, respectively. From proximal to distal: the vascular axis includes vascular smooth muscle cells and pericytes that transition as arterioles and venules ramify into capillaries; the epithelial axis includes airway smooth muscle cells and two populations of myofibroblasts - ductal myofibroblasts, surrounding alveolar ducts and marked by CDH4, HHIP and LGR6, which persist post-alveologenesis, and alveolar myofibroblasts, surrounding alveoli and marked by high expression of PDGFRA, which undergo developmental apoptosis; and the interstitial axis, residing between the epithelial and vascular trees and sharing the marker MEOX2, includes fibroblasts in the bronchovascular bundle and the alveolar interstitium, which are marked by IL33/DNER/PI16 and Wnt2, respectively. Single-cell imaging reveals a distinct morphology of mesenchymal cell populations. This classification provides a conceptual and experimental framework applicable to other organs.
Collapse
Affiliation(s)
- Odemaris Narvaez Del Pilar
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Graduate School of Biomedical Sciences , The University of Texas MD Anderson Cancer Center UTHealth, Houston, Texas 77030, USA.,University of Puerto Rico - Medical Sciences Campus, San Juan, Puerto Rico 00927
| | - Maria Jose Gacha Garay
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Graduate School of Biomedical Sciences , The University of Texas MD Anderson Cancer Center UTHealth, Houston, Texas 77030, USA
| | - Jichao Chen
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
4
|
Muhiddin HS, Kamaruddin MI, Ichsan AM, Budu. Vitreous and Serum Concentrations of Vascular Endothelial Growth Factor and Platelet-Derived Growth Factor in Proliferative Diabetic Retinopathy. Clin Ophthalmol 2020; 14:1547-1552. [PMID: 32606568 PMCID: PMC7293416 DOI: 10.2147/opth.s248812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/18/2020] [Indexed: 01/04/2023] Open
Abstract
Purpose This study aimed to investigate the concentrations of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) in vitreous and serum samples, analyze the ratio, and compare among proliferative diabetic retinopathy (PDR) subgroups. Patients and Methods This study included 17 eyes of patients with PDR, identified as the PDR group which was divided into three subgroups (vitreous hemorrhage [VH], VH with fibrotic tissues, and tractional retinal detachment), and five control eyes (nucleus and intraocular lens drop). Vitreous and serum samples were obtained on the same day. The VEGF-A and PDGF-AB concentrations were calculated by enzyme-linked immunosorbent assay. Results The VEGF-A and PDGF-AB concentrations in vitreous samples were significantly higher in the PDR group (630.72 ± 342.81 pg/mL) compared with those in the control group (153.58 ± 145.85 pg/mL); however, they were not detected in serum samples. The vitreous/serum ratio of the VEGF-A concentration in the PDR group (2.1 ± 1.8) was significantly higher compared with that in the control group (0.31 ± 0.33). The VEGF-A concentrations in vitreous samples were highest in the VH group and lowest in the VH with fibrotic tissue subgroup (mean difference 536.16 pg/mL). The vitreous VEGF-A/PDGF-AB concentration ratios were also significantly different among the PDR subgroups. Conclusion High concentrations of VEGF and PDGF in vitreous samples of PDR eyes indicate its local related activity in PDR pathology. There is a possibility of PDGF involvement in the pathogenesis of PDR. The VEGF/PDGF concentration ratios possibly play a significant role in the formation of fibrotic tissue in PDR.
Collapse
Affiliation(s)
- Habibah Setyawati Muhiddin
- Department of Ophthalmology, Faculty of Medicine, Hasanuddin University, Makassar, South Sulawesi, Indonesia
| | - Muhammad Irfan Kamaruddin
- Department of Ophthalmology, Faculty of Medicine, Hasanuddin University, Makassar, South Sulawesi, Indonesia
| | - Andi Muhammad Ichsan
- Department of Ophthalmology, Faculty of Medicine, Hasanuddin University, Makassar, South Sulawesi, Indonesia
| | - Budu
- Department of Ophthalmology, Faculty of Medicine, Hasanuddin University, Makassar, South Sulawesi, Indonesia
| |
Collapse
|
5
|
Maleknia M, Ansari N, Haybar H, Maniati M, Saki N. Inflammatory Growth Factors and In-Stent Restenosis: Effect of Cytokines and Growth Factors. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s42399-020-00240-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
6
|
Sun T, Yin L, Kuang H. miR-181a/b-5p regulates human umbilical vein endothelial cell angiogenesis by targeting PDGFRA. Cell Biochem Funct 2019; 38:222-230. [PMID: 31879991 DOI: 10.1002/cbf.3472] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/19/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a growing burden in low-and middle-income countries. Changing lifestyles and lack of physical activity are some of the reasons contributing to this epidemic increase. Co-morbidities associated with T2DM are largely due to the complications which arise as a consequence of endothelial dysfunction. Platelet derived growth factor-alpha (PDGFRA) is a protein responsible for cell proliferation, angiogenesis, migration and invasion. Increased levels of PDGFRA have been reported in T2DM. This study assessed the epigenetic regulation of PDGFRA through microRNAs (miR-181a/b-5p).Using a bioinformatics-based approach, we assessed the binding of miR-181a/b-5p to PDGFRA. Experimentally, this binding was confirmed using a dual luciferase reporter assay. Further, we overexpressed miR-181a/b-5p in Human umbilical vein endothelial cells (HUVECs) and the influence of over-expression on cell proliferation, migration and angiogenesis was assessed using in-vitro approaches. The influence of miR-181a/b-5p over expression on cellular apoptosis was ascertained using a TUNEL assay with concomitant changes being observed in the levels of Bcl-2 and cleaved Caspase-3.In HUVECs, PDGFRA is a direct target for miR-181a/b-5p. Over expression of miR-181a/b-5p decreased cellular proliferation, migration, invasion, and tube formation-a surrogate marker for angiogenesis. miR-181a/b-5p may be used as a therapeutic intervention to restrict uncontrolled levels of PDGFRA and thereby rescue the phenotypes of increased cell proliferation, migration, invasion and tube formation. miR-181a/b negatively regulates PDGFRA levels. Significance of the study: T2DM and its associated complications emerge from endothelial dysfunction. The associated phenotypes are regulated by a number of proteins, one such member being, PDGFRA. PDGFRA is in turn regulated by miR-181a/b-5p. Complementation with miR-181a/b-5p resulted in reversion of phenotypes. Thus, miR-181a/b-5p-mediated suppression of PDGFRA may be used as a therapeutic intervention in the management of type 2 diabetes.
Collapse
Affiliation(s)
- Tingting Sun
- Department of Gerontology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linan Yin
- Department of Interventional Radiology, Harbin Medical University Cancer hospital, Harbin, China
| | - Hongyu Kuang
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
7
|
Liu B, Song J, Han H, Hu Z, Chen N, Cui J, Matsubara JA, Zhong J, Lei H. Blockade of MDM2 with inactive Cas9 prevents epithelial to mesenchymal transition in retinal pigment epithelial cells. J Transl Med 2019; 99:1874-1886. [PMID: 31439892 DOI: 10.1038/s41374-019-0307-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 01/10/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) plays an important role in the pathogenesis of proliferative vitreoretinopathy (PVR). We aimed to demonstrate the role of mouse double minute 2 (MDM2) in transforming growth factor-beta 2 (TGF-β2)-induced EMT in human retinal pigment epithelial cells (RPEs). Immunofluorescence was used to assess MDM2 expression in epiretinal membranes (ERMs) from patients with PVR. A single guide (sg)RNA targeting the second promoter of MDM2 was cloned into a mutant lentiviral Clustered Regularly Interspaced Short Palindromic Repeats (lentiCRISPR) v2 (D10A and H840A) vector for expressing nuclease dead Cas9 (dCas9)/MDM2-sgRNA in RPEs. In addition, MDM2-sgRNA was also cloned into a pLV-sgRNA-dCas9-Kruppel associated box (KRAB) vector for expressing dCas9 fused with a transcriptional repressor KRAB/MDM2-sgRNA. TGF-β2-induced expression of MDM2 and EMT biomarkers were assessed by quantitative polymerase chain reaction (q-PCR), western blot, or immunofluorescence. Wound-healing and proliferation assays were used to evaluate the role of MDM2 in TGF-β2-induced responses in RPEs. As a result, we found that MDM2 was expressed obviously in ERMs, and that TGF-β2-induced expression of MDM2 and EMT biomarkers Fibronectin, N-cadherin and Vimentin in RPEs. Importantly, we discovered that the dCas9/MDM2-sgRNA blocked TGF-β2-induced expression of MDM2 and the EMT biomarkers without affecting their basal expression, whereas the dCas9-KRAB/MDM2-sgRNA suppressed basal MDM2 expression in RPEs. These cells could not be maintained continuously because their viability was greatly reduced. Next, we found that Nutlin-3, a small molecule blocking the interaction of MDM2 with p53, inhibited TGF-β2-induced expression of Fibronectin and N-cadherin but not Vimentin in RPEs, indicating that MDM2 functions in both p53-dependent and -independent pathways. Finally, our experimental data demonstrated that dCas9/MDM2-sgRNA suppressed TGF-β2-dependent cell proliferation and migration without disturbing the unstimulated basal activity. In conclusion, the CRISPR/dCas9 capability for blocking TGF-β2-induced expression of MDM2 and EMT biomarkers can be exploited for a therapeutic approach to PVR.
Collapse
Affiliation(s)
- Bing Liu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, China
| | - Jingyuan Song
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, 100193, Beijing, China
| | - Haote Han
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Department of Biomedical Engineering, Zhejiang University, 310027, Hangzhou, China
| | - Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Na Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Renji Hospital School of Medicine, Shanghai Jiaotong University, 200127, Shanghai, China
| | - Jing Cui
- The University of British Columbia, Vancouver, BC, V5Z 3N9, Canada
| | | | - Jingxiang Zhong
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, China
| | - Hetian Lei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
8
|
Chen S, Imoukhuede PI. Multiplexing Angiogenic Receptor Quantification via Quantum Dots. Anal Chem 2019; 91:7603-7612. [DOI: 10.1021/acs.analchem.9b00238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Si Chen
- Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, Missouri 63130, United States
| | - P. I. Imoukhuede
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, Missouri 63130, United States
| |
Collapse
|
9
|
Chen S, Le T, Harley BAC, Imoukhuede PI. Characterizing Glioblastoma Heterogeneity via Single-Cell Receptor Quantification. Front Bioeng Biotechnol 2018; 6:92. [PMID: 30050899 PMCID: PMC6050407 DOI: 10.3389/fbioe.2018.00092] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/21/2018] [Indexed: 01/09/2023] Open
Abstract
Dysregulation of tyrosine kinase receptor (RTK) signaling pathways play important roles in glioblastoma (GBM). However, therapies targeting these signaling pathways have not been successful, partially because of drug resistance. Increasing evidence suggests that tumor heterogeneity, more specifically, GBM-associated stem and endothelial cell heterogeneity, may contribute to drug resistance. In this perspective article, we introduce a high-throughput, quantitative approach to profile plasma membrane RTKs on single cells. First, we review the roles of RTKs in cancer. Then, we discuss the sources of cell heterogeneity in GBM, providing context to the key cells directing resistance to drugs. Finally, we present our provisionally patented qFlow cytometry approach, and report results of a "proof of concept" patient-derived xenograft GBM study.
Collapse
Affiliation(s)
- Si Chen
- Department of Bioengineering, University of Illinois at Urbana–Champaign, Champaign, IL, United States
| | - Thien Le
- Department of Mathematics and Department of Computer Science, University of Illinois at Urbana–Champaign, Champaign, IL, United States
| | - Brendan A. C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana–Champaign, Urbana, IL, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - P. I. Imoukhuede
- Department of Bioengineering, University of Illinois at Urbana–Champaign, Champaign, IL, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
| |
Collapse
|
10
|
Wen Z, Shen Y, Berry G, Shahram F, Li Y, Watanabe R, Liao YJ, Goronzy JJ, Weyand CM. The microvascular niche instructs T cells in large vessel vasculitis via the VEGF-Jagged1-Notch pathway. Sci Transl Med 2018; 9:9/399/eaal3322. [PMID: 28724574 DOI: 10.1126/scitranslmed.aal3322] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/07/2017] [Accepted: 05/31/2017] [Indexed: 12/11/2022]
Abstract
Microvascular networks in the adventitia of large arteries control access of inflammatory cells to the inner wall layers (media and intima) and thus protect the immune privilege of the aorta and its major branches. In autoimmune vasculitis giant cell arteritis (GCA), CD4 T helper 1 (TH1) and TH17 cells invade into the wall of the aorta and large elastic arteries to form tissue-destructive granulomas. Whether the disease microenvironment provides instructive cues for vasculitogenic T cells is unknown. We report that adventitial microvascular endothelial cells (mvECs) perform immunoregulatory functions by up-regulating the expression of the Notch ligand Jagged1. Vascular endothelial growth factor (VEGF), abundantly present in GCA patients' blood, induced Jagged1 expression, allowing mvECs to regulate effector T cell induction via the Notch-mTORC1 (mammalian target of rapamycin complex 1) pathway. We found that circulating CD4 T cells in GCA patients have left the quiescent state, actively signal through the Notch pathway, and differentiate into TH1 and TH17 effector cells. In an in vivo model of large vessel vasculitis, exogenous VEGF functioned as an effective amplifier to recruit and activate vasculitogenic T cells. Thus, systemic VEGF co-opts endothelial Jagged1 to trigger aberrant Notch signaling, biases responsiveness of CD4 T cells, and induces pathogenic effector functions. Adventitial microvascular networks function as an instructive tissue niche, which can be exploited to target vasculitogenic immunity in large vessel vasculitis.
Collapse
Affiliation(s)
- Zhenke Wen
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yi Shen
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gerald Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Farhad Shahram
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yinyin Li
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ryu Watanabe
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yaping Joyce Liao
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
11
|
Stangret A, Skoda M, Wnuk A, Pyzlak M, Szukiewicz D. Mild anemia during pregnancy upregulates placental vascularity development. Med Hypotheses 2017; 102:37-40. [PMID: 28478827 DOI: 10.1016/j.mehy.2017.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/05/2017] [Indexed: 11/16/2022]
Abstract
The connection between maternal hematological status and pregnancy outcome has been shown by many independent researchers. Attention was initially focused on the adverse effects of moderate and severe anemia. Interestingly, some studies revealed that mild anemia was associated with optimal fetal development and was not affecting pregnancy outcome. The explanation for this phenomenon became a target for scientists. Hemodilution, physiologic anemia and relative decrease in hemoglobin concentration are the changes observed during pregnancy but they do not explain the reasons for the positive influence of mild anemia on a fetomaternal unit. It is hypothesized that hemodilution facilitates placental perfusion because blood viscosity is reduced. Subsequently, it may lead to a decline in hemoglobin concentration. Anemia from its definition implies decreased oxygen carrying capacity of the blood and can result in hypoxemia and even hypoxia, which is a common factor inducing new blood vessels formation. Therefore, we raised the hypothesis that the lowered hemoglobin concentration during pregnancy may upregulate vascular growth factor receptors expression such as VEGFR-1 (Flt-1) and VEGFR-2 (FLK-1/KDR). Consecutively, increased fetoplacental vasculogenesis and angiogenesis provide further expansion of vascular network development, better placental perfusion and hence neither fetus nor the mother are affected.
Collapse
Affiliation(s)
- A Stangret
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland.
| | - M Skoda
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland
| | - A Wnuk
- Chair and Department of Obstetrics, Gynecology, and Oncology, 2nd Faculty of Medical University of Warsaw, Poland
| | - M Pyzlak
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland
| | - D Szukiewicz
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland
| |
Collapse
|
12
|
Kazlauskas A. PDGFs and their receptors. Gene 2017; 614:1-7. [PMID: 28267575 DOI: 10.1016/j.gene.2017.03.003] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 01/28/2023]
Abstract
The platelet-derived growth factor (PDGF)/PDGFR receptor (PDGFR) family is essential for a vast array of physiological processes such as migration and proliferation of percityes that contribute to the formation and proper function of blood vessels. While ligand-dependent de-repression of the PDGFR's kinase activity is the major mode by which the PDGFR is activated, there are additional mechanisms to activate PDGFRs. Deregulated PDGFR activity contributes to various pathological conditions, and hence the PDGF/PDGFR family members are viable therapeutic targets. An increased appreciation of which PDGFR contributes to pathology, biomarkers that indicate the amplitude and mode of activation, and receptor-specific antagonists are necessary for the development of next-generation therapies that target the PDGF/PDGFR family.
Collapse
Affiliation(s)
- Andrius Kazlauskas
- Schepens Eye Research Institute, Massachusetts Eye and Ear Institute, 20 Staniford St, Boston, MA 02114, United States.
| |
Collapse
|