1
|
Chaudhari PJ, Nemade AR, Shirkhedkar AA. Recent updates on potential of VEGFR-2 small-molecule inhibitors as anticancer agents. RSC Adv 2024; 14:33384-33417. [PMID: 39439843 PMCID: PMC11495155 DOI: 10.1039/d4ra05244g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
The vascular endothelial growth factor receptor (VEGFR) system is the key component for controlling angiogenesis in cancer cells. Blocking vascular endothelial growth factor receptor 2 (VEGFR2) signalling is one of the most promising approaches to hindering angiogenesis and the subsequent growth of cancer cells. The USFDA-approved small-molecule drugs targeting VEGFR-2 are developing drug resistance over the course of chemotherapy, and cardiac-related side effects are consistently being reported; hence, there is an urgent need for more safe and effective anticancer molecules. The present review focuses on the structure and physiology of VEGFR-2 and its involvement in the progression of cancer cells. The recent updates from the last five years through papers and patents on structure-activity relationships, pharmacophoric attributes, molecular docking interactions, antiangiogenic assays, cancer cell line studies, and the potencies (IC50) of VEGFR-2 inhibitors are discussed herein. The common structural framework requirements, such as the Asp-Phe-Gly (DFG) motif of VEGFR-2 interacting with the HBD-HBA region in the ligand molecules, the central aryl ring occupying the linker region, and a variety of bio-isosteres, can enhance activity against VEGFR-2. At one end, the heteroaryl moiety is essential for interaction within the ATP-binding site of VEGFR-2, while the terminal hydrophobic tail occupies the allosteric binding site. Three to five bond spacers between the heteroaryl and HBD-HBA regions provided a better result towards VEGFR-2 inhibition, mirroring the behaviors of standard drugs. The in-depth analysis of recent updates on VEGFR-2 inhibitors presented in this paper will help prospective synthetic and medicinal chemists to discover new lead molecules for the treatment of various cancers.
Collapse
Affiliation(s)
- Prashant Jagannath Chaudhari
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research Shirpur, Dist-Dhule Maharashtra 425 405 India
- Department of Chemistry, Carnegie Mellon University 4400 Fifth Avenue Pittsburgh Pennsylvania 15213 USA
| | - Aditya Ramchandra Nemade
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research Shirpur, Dist-Dhule Maharashtra 425 405 India
- Department of Pharmaceutics, M.S. Ramaiah University of Applied Sciences Bengaluru Karnataka 560054 India
| | - Atul Arun Shirkhedkar
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research Shirpur, Dist-Dhule Maharashtra 425 405 India
| |
Collapse
|
2
|
Oguri G, Ikegami R, Ugawa H, Katoh M, Obi S, Sakuma M, Takeda N, Kano Y, Toyoda S, Nakajima T. Muscle Atrophy and mRNA-miRNA Network Analysis of Vascular Endothelial Growth Factor (VEGF) in a Mouse Model of Denervation-Induced Disuse. Cureus 2024; 16:e68974. [PMID: 39385898 PMCID: PMC11462388 DOI: 10.7759/cureus.68974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Skeletal muscle atrophy is frequently caused by the disuse of muscles. It impacts quality of life, especially in aging populations and those with chronic diseases. Understanding the molecular mechanisms underlying muscle atrophy is crucial for developing effective therapies. OBJECTIVE To investigate the roles of vascular endothelial growth factor (VEGF) and various microRNAs (miRNAs) in muscle atrophy using a mouse model of denervation (DEN)-induced disuse, and to elucidate their interactions and regulatory functions through comprehensive network analysis. METHODS The right sciatic nerve of C57BL/6J mice (n=6) was excised to simulate DEN, with the left serving as a sham surgery control (Sham). Following a two-week period, wet muscle weight was measured. Total RNA was extracted from the tibialis anterior muscle for microarray analysis. Significant expression changes were analyzed via Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and miRNet for miRNAs. RESULTS Denervated limbs showed a significant reduction in muscle weight. Over 1,000 genes displayed increased expression, while 527 showed reductions to less than half of control levels. VEGF, along with specific miRNAs such as miR-106a-5p, miR-mir20a-5p, mir93-5p and mir17-5p, occupied central regulatory nodes within the gene network. Functional analysis revealed that these molecules are involved in key biological processes including regulation of cell migration, vasculature development, and regulation of endothelial cell proliferation. The increased miRNAs were subjected to further network analysis that revealed significant regulatory interactions with target mRNAs. CONCLUSION VEGF and miRNAs play crucial roles in the progression of skeletal muscle atrophy, offering potential targets for therapeutic interventions aimed at reducing atrophy and enhancing muscle regeneration.
Collapse
Affiliation(s)
- Gaku Oguri
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, JPN
| | - Ryo Ikegami
- Department of Information Science and Technology, The University of Electro-Communications, Tokyo, JPN
- Department of Physical Therapy, Niigata University of Health and Welfare, Niigata, JPN
| | - Haruka Ugawa
- Department of Information Science and Technology, The University of Electro-Communications, Tokyo, JPN
- Department of Physical Therapy, Niigata University of Health and Welfare, Niigata, JPN
| | - Manami Katoh
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, JPN
| | - Syotaro Obi
- Department of Cardiovascular Medicine, Dokkyo Medical University Hospital, Mibu, JPN
| | - Masashi Sakuma
- Department of Cardiovascular Medicine, Dokkyo Medical University Hospital, Mibu, JPN
| | - Norihiko Takeda
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, JPN
| | - Yutaka Kano
- Department of Information Science and Technology, The University of Electro-Communications, Tokyo, JPN
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University Hospital, Mibu, JPN
| | - Toshiaki Nakajima
- Department of Cardiovascular Medicine, Dokkyo Medical University Hospital, Mibu, JPN
- Department of Medical KAATSU Training, Dokkyo Medical University Hospital, Mibu, JPN
| |
Collapse
|
3
|
Tai Y, Chow A, Han S, Coker C, Ma W, Gu Y, Estrada Navarro V, Kandpal M, Hibshoosh H, Kalinsky K, Manova-Todorova K, Safonov A, Walsh EM, Robson M, Norton L, Baer R, Merghoub T, Biswas AK, Acharyya S. FLT1 activation in cancer cells promotes PARP-inhibitor resistance in breast cancer. EMBO Mol Med 2024; 16:1957-1980. [PMID: 38956205 PMCID: PMC11319505 DOI: 10.1038/s44321-024-00094-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Acquired resistance to PARP inhibitors (PARPi) remains a treatment challenge for BRCA1/2-mutant breast cancer that drastically shortens patient survival. Although several resistance mechanisms have been identified, none have been successfully targeted in the clinic. Using new PARPi-resistance models of Brca1- and Bard1-mutant breast cancer generated in-vivo, we identified FLT1 (VEGFR1) as a driver of resistance. Unlike the known role of VEGF signaling in angiogenesis, we demonstrate a novel, non-canonical role for FLT1 signaling that protects cancer cells from PARPi in-vivo through a combination of cell-intrinsic and cell-extrinsic pathways. We demonstrate that FLT1 blockade suppresses AKT activation, increases tumor infiltration of CD8+ T cells, and causes dramatic regression of PARPi-resistant breast tumors in a T-cell-dependent manner. Moreover, PARPi-resistant tumor cells can be readily re-sensitized to PARPi by targeting Flt1 either genetically (Flt1-suppression) or pharmacologically (axitinib). Importantly, a retrospective series of breast cancer patients treated with PARPi demonstrated shorter progression-free survival in cases with FLT1 activation at pre-treatment. Our study therefore identifies FLT1 as a potential therapeutic target in PARPi-resistant, BRCA1/2-mutant breast cancer.
Collapse
Affiliation(s)
- Yifan Tai
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Biology, McGill University, Montreal, Quebec, QC, H3G0B1, Canada
| | - Angela Chow
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Seoyoung Han
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Jacobs School of Medicine, University of Buffalo, New York, NY, USA
| | - Courtney Coker
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Wanchao Ma
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Yifan Gu
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Valeria Estrada Navarro
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Manoj Kandpal
- Centre for Clinical and Translational Science, Rockefeller University Hospital, 1230 York Ave, New York, NY, 10065, USA
| | - Hanina Hibshoosh
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Kevin Kalinsky
- Winship Cancer Institute of Emory University, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA, 30322, USA
| | - Katia Manova-Todorova
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Anton Safonov
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Elaine M Walsh
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Department of Medicine, Georgetown Lombardi Comprehensive Cancer Center, 3800 Reservoir Rd, NW, Washington DC, 20007, USA
| | - Mark Robson
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Larry Norton
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Richard Baer
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Taha Merghoub
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Anup K Biswas
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Swarnali Acharyya
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, 1130 St. Nicholas Ave, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
4
|
North HF, Weissleder C, Bitar M, Barry G, Fullerton JM, Webster MJ, Weickert CS. RNA-sequencing suggests extracellular matrix and vasculature dysregulation could impair neurogenesis in schizophrenia cases with elevated inflammation. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:50. [PMID: 38704390 PMCID: PMC11069512 DOI: 10.1038/s41537-024-00466-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/25/2024] [Indexed: 05/06/2024]
Abstract
A subgroup of schizophrenia cases with elevated inflammation have reduced neurogenesis markers and increased macrophage density in the human subependymal zone (SEZ; also termed subventricular zone or SVZ) neurogenic niche. Inflammation can impair neurogenesis; however, it is unclear which other pathways are associated with reduced neurogenesis. This research aimed to discover transcriptomic differences between inflammatory subgroups of schizophrenia in the SEZ. Total RNA sequencing was performed on SEZ tissue from schizophrenia cases, designated into low inflammation (n = 13) and high inflammation (n = 14) subgroups, based on cluster analysis of inflammation marker gene expression. 718 genes were differentially expressed in high compared to low inflammation schizophrenia (FDR p < 0.05) and were most significantly over-represented in the pathway 'Hepatic Fibrosis/Hepatic Stellate-Cell Activation'. Genes in this pathway relate to extracellular matrix stability (including ten collagens) and vascular remodelling suggesting increased angiogenesis. Collagen-IV, a key element of the basement membrane and fractones, had elevated gene expression. Immunohistochemistry revealed novel collagen-IV+ fractone bulbs within the human SEZ hypocellular gap. Considering the extracellular matrix's regulatory role in SEZ neurogenesis, fibrosis-related alterations in high inflammation schizophrenia may disrupt neurogenesis. Increased angiogenesis could facilitate immune cell transmigration, potentially explaining elevated macrophages in high inflammation schizophrenia. This discovery-driven analysis sheds light on how inflammation may contribute to schizophrenia neuropathology in the neurogenic niche.
Collapse
Affiliation(s)
- Hayley F North
- Neuroscience Research Australia, Sydney, NSW, Australia
- Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Christin Weissleder
- Neuroscience Research Australia, Sydney, NSW, Australia
- Mechanism and therapy for genetic brain diseases, Institut Imagine, Paris, France
| | | | - Guy Barry
- OncoLife Therapeutics, Yeronga, QLD, Australia
| | - Janice M Fullerton
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, 9800, Medical Center Drive, Rockville, MD, USA
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia, Sydney, NSW, Australia.
- Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
5
|
Huang XL, Sun Y, Wen P, Pan JC, He WY. The potential mechanism of ursolic acid in the treatment of bladder cancer based on network pharmacology and molecular docking. J Int Med Res 2024; 52:3000605241234006. [PMID: 38443785 PMCID: PMC10916484 DOI: 10.1177/03000605241234006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
OBJECTIVE This study explored the potential molecular mechanisms of ursolic acid (UA) in bladder cancer treatment using network pharmacology and molecular docking. METHODS The Traditional Chinese Medicine Systems Pharmacology and UniProt databases were used to screen potential targets of UA. Relevant bladder cancer target genes were extracted using the GeneCards database. All data were pooled and intercrossed to obtain common target genes of UA and bladder cancer. Gene Ontology functional annotation and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed. Molecular docking was conducted to verify the possible binding conformation between UA and bladder cancer cells. Then, in vitro experiments were performed to further validate the predicted results. RESULTS UA exerts anti-tumor effects on bladder cancer through multiple targets and pathways. Molecular docking indicated that UA undergoes stable binding with the proteins encoded by the top six core genes (STAT3, VEGFA, CASP3, TP53, IL1B, and CCND1). The in vitro experiments verified that UA can induce bladder cancer cell apoptosis by regulating the PI3K/Akt signaling pathway. CONCLUSIONS Our study illustrated the potential mechanism of UA in bladder cancer based on network pharmacology and molecular docking. The results will provide scientific references for follow-up studies and clinical treatment.
Collapse
Affiliation(s)
- Xiao-Long Huang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Urology, People’s Hospital of Hechuan, Chongqing, China
| | - Yan Sun
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Wen
- Department of Urology, People’s Hospital of Hechuan, Chongqing, China
| | - Jun-Cheng Pan
- Department of Urology, People’s Hospital of Hechuan, Chongqing, China
| | - Wei-Yang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Takenoshita Y, Tokito A, Jougasaki M. Inhibitory Effects of Eicosapentaenoic Acid on Vascular Endothelial Growth Factor-Induced Monocyte Chemoattractant Protein-1, Interleukin-6, and Interleukin-8 in Human Vascular Endothelial Cells. Int J Mol Sci 2024; 25:2749. [PMID: 38473995 PMCID: PMC10931732 DOI: 10.3390/ijms25052749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Vascular endothelial growth factor (VEGF) induces monocyte chemoattractant protein-1 (MCP-1) and plays an important role in vascular inflammation and atherosclerosis. We investigated the mechanisms of VEGF-induced MCP-1 expression and the effects of eicosapentaenoic acid (EPA) in human umbilical vein endothelial cells (HUVECs). Real-time reverse transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) demonstrated that VEGF enhanced MCP-1 gene expression and protein secretion in HUVECs. Western immunoblot analysis revealed that VEGF induced the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and inhibitor of nuclear factor (NF)-κB (IκB). Treatment with pharmacological inhibitors of p38 MAPK (SB203580) or NF-κB (BAY11-7085) significantly suppressed VEGF-induced MCP-1 in HUVECs. EPA inhibited VEGF-induced MCP-1 mRNA, protein secretion, phosphorylation of p38 MAPK, and the translocation of phospho-p65 to the nucleus. Additionally, VEGF also stimulated gene expressions of interleukin (IL)-6 and IL-8, which were suppressed by SB203580, BAY11-7085, and EPA. The present study has demonstrated that VEGF-induced activation of MCP-1, IL-6, and IL-8 involves the p38 MAPK and NF-κB signaling pathways and that EPA inhibits VEGF-induced MCP-1, IL-6, and IL-8 via suppressing these signaling pathways. This study supports EPA as a beneficial anti-inflammatory and anti-atherogenic drug to reduce the VEGF-induced activation of proinflammatory cytokine and chemokines.
Collapse
Affiliation(s)
| | | | - Michihisa Jougasaki
- Institute for Clinical Research, NHO Kagoshima Medical Center, Kagoshima 892-0853, Japan; (Y.T.); (A.T.)
| |
Collapse
|
7
|
Ceylan HK, Kırbay FÖ, Yazgan İ, Elibol M. A colorimetric immunoassay for the detection of human vascular endothelial growth factor 165 (VEGF 165) based on anti-VEGF-iron oxide nanoparticle conjugation. Mikrochim Acta 2024; 191:133. [PMID: 38353782 PMCID: PMC10867064 DOI: 10.1007/s00604-024-06228-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Vascular endothelial growth factor (VEGF) is an indispensable element in many physiological processes, while alterations in its level in the circulating system are signs of pathology-associated diseases. Therefore, its precise and selective detection is critical for clinical applications to monitor the progression of the pathology. In this study, an optical immunoassay biosensor was developed as a model study for detecting recombinant VEGF165. The VEGF165 sample was purified from recombinant Kluyveromyces lactis GG799 yeast cells. Indirect ELISA was used during the detection, wherein iron oxide nanoparticles (FeNPs) were utilized to obtain optical signals. The FeNPs were synthesized in the presence of lactose p-amino benzoic acid (LpAB). VEGF165 antibody was conjugated to the LpAB-FeNPs through EDC/NHS chemistry to convert the iron oxide nanoparticles into VEGF165 specific probes. The specificity of the prepared system was tested in the presence of potential serum-based interferents (i.e., glucose, urea, insulin, C-reactive protein, and serum amyloid A), and validation studies were performed in a simulated serum sample. The proposed immunoassay showed a wide detection range (0.5 to 100 ng/mL) with a detection limit of 0.29 ng/mL. These results show that the developed assay could offer a sensitive, simple, specific, reliable, and high-throughput detection platform that can be used in the clinical diagnostics of VEGF.
Collapse
Affiliation(s)
- Hülya Kuduğ Ceylan
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Tokat Gaziosmanpaşa University, 60250, Tokat, Turkey.
| | - Fatma Öztürk Kırbay
- Biochemistry Department, Faculty of Science, Ege University, Bornova, 35100, Izmir, Turkey.
| | - İdris Yazgan
- Center for Biosensors and Material Science, Department of Biology, Faculty of Science and Art, Kastamonu University, 37100, Kastamonu, Turkey
| | - Murat Elibol
- Bioengineering Department, Ege University, Bornova, 35100, Izmir, Turkey
| |
Collapse
|
8
|
Stachyra-Strawa P, Szatkowska-Sieczek L, Cisek P, Gołębiowski P, Grzybowska-Szatkowska L. Cardiac and Nephrological Complications Related to the Use of Antiangiogenic and Anti-Programmed Cell Death Protein 1 Receptor/Programmed Cell Death Protein 1 Ligand Therapy. Genes (Basel) 2024; 15:177. [PMID: 38397167 PMCID: PMC10887630 DOI: 10.3390/genes15020177] [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: 12/10/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The ability to undergo neoangiogenesis is a common feature with all cancers. Signaling related to vascular endothelial growth factors (VEGF) and their receptors (VEGFR) plays a key role in the process of tumor neoangiogenesis. A close relationship has been demonstrated between excessive VEGF levels and the induction of immunosuppression in the tumor microenvironment. The use of drugs blocking the VEGF function, apart from the anticancer effect, also result in adverse effects, in particular related to the circulatory system and kidneys. Cardiac toxicity associated with the use of such therapy manifests itself mainly in the form of hypertension, thromboembolic episodes and ischemic heart disease. In the case of renal complications, the most common symptoms include renal arterial hypertension, proteinuria and microangiopathy. Although these complications are reversible in 60-80% of cases after cessation of VSP (VEGF pathway inhibitor) therapy, in some cases they can lead to irreversible changes in renal function, whereas cardiac complications may be fatal. Also, the use of PD-1/PD-L1 inhibitors may result in kidney and heart damage. In the case of cardiac complications, the most common symptoms include myocarditis, pericarditis, arrhythmia, acute coronary syndrome and vasculitis, while kidney damage most often manifests as acute kidney injury (AKI), nephrotic syndrome, pyuria or hematuria. The decision whether to resume treatment after the occurrence of cardiovascular and renal complications remains a problem.
Collapse
Affiliation(s)
- Paulina Stachyra-Strawa
- Department of Radiotherapy, Medical University of Lublin, Chodźki 7, 20-093 Lublin, Poland; (P.S.-S.); (P.C.); (P.G.)
| | - Lidia Szatkowska-Sieczek
- Clinical Department of Cardiology, 4th Military Hospital, Rudolfa Weigla 5, 50-981 Wroclaw, Poland;
| | - Paweł Cisek
- Department of Radiotherapy, Medical University of Lublin, Chodźki 7, 20-093 Lublin, Poland; (P.S.-S.); (P.C.); (P.G.)
| | - Paweł Gołębiowski
- Department of Radiotherapy, Medical University of Lublin, Chodźki 7, 20-093 Lublin, Poland; (P.S.-S.); (P.C.); (P.G.)
| | - Ludmiła Grzybowska-Szatkowska
- Department of Radiotherapy, Medical University of Lublin, Chodźki 7, 20-093 Lublin, Poland; (P.S.-S.); (P.C.); (P.G.)
| |
Collapse
|
9
|
Chen TA, Sharma D, Jia W, Ha D, Man K, Zhang J, Yang Y, Zhou Y, Kamp TJ, Zhao F. Detergent-Based Decellularization for Anisotropic Cardiac-Specific Extracellular Matrix Scaffold Generation. Biomimetics (Basel) 2023; 8:551. [PMID: 37999192 PMCID: PMC10669368 DOI: 10.3390/biomimetics8070551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/19/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
Cell-derived extracellular matrix (ECM) has become increasingly popular in tissue engineering applications due to its ability to provide tailored signals for desirable cellular responses. Anisotropic cardiac-specific ECM scaffold decellularized from human induced pluripotent stem cell (hiPSC)-derived cardiac fibroblasts (hiPSC-CFs) mimics the native cardiac microenvironment and provides essential biochemical and signaling cues to hiPSC-derived cardiomyocytes (hiPSC-CMs). The objective of this study was to assess the efficacy of two detergent-based decellularization methods: (1) a combination of ethylenediaminetetraacetic acid and sodium dodecyl sulfate (EDTA + SDS) and (2) a combination of sodium deoxycholate and deoxyribonuclease (SD + DNase), in preserving the composition and bioactive substances within the aligned ECM scaffold while maximumly removing cellular components. The decellularization effects were evaluated by characterizing the ECM morphology, quantifying key structural biomacromolecules, and measuring preserved growth factors. Results showed that both treatments met the standard of cell removal (less than 50 ng/mg ECM dry weight) and substantially preserved major ECM biomacromolecules and growth factors. The EDTA + SDS treatment was more time-efficient and has been determined to be a more efficient method for generating an anisotropic ECM scaffold from aligned hiPSC-CFs. Moreover, this cardiac-specific ECM has demonstrated effectiveness in supporting the alignment of hiPSC-CMs and their expression of mature structural and functional proteins in in vitro cultures, which is crucial for cardiac tissue engineering.
Collapse
Affiliation(s)
- Te-An Chen
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Dhavan Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Wenkai Jia
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Donggi Ha
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Jianhua Zhang
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Yuxiao Zhou
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Timothy J. Kamp
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
10
|
Rabah N, Ait Mohand FE, Kravchenko-Balasha N. Understanding Glioblastoma Signaling, Heterogeneity, Invasiveness, and Drug Delivery Barriers. Int J Mol Sci 2023; 24:14256. [PMID: 37762559 PMCID: PMC10532387 DOI: 10.3390/ijms241814256] [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: 08/29/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The most prevalent and aggressive type of brain cancer, namely, glioblastoma (GBM), is characterized by intra- and inter-tumor heterogeneity and strong spreading capacity, which makes treatment ineffective. A true therapeutic answer is still in its infancy despite various studies that have made significant progress toward understanding the mechanisms behind GBM recurrence and its resistance. The primary causes of GBM recurrence are attributed to the heterogeneity and diffusive nature; therefore, monitoring the tumor's heterogeneity and spreading may offer a set of therapeutic targets that could improve the clinical management of GBM and prevent tumor relapse. Additionally, the blood-brain barrier (BBB)-related poor drug delivery that prevents effective drug concentrations within the tumor is discussed. With a primary emphasis on signaling heterogeneity, tumor infiltration, and computational modeling of GBM, this review covers typical therapeutic difficulties and factors contributing to drug resistance development and discusses potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, Hebrew University of Jerusalem, Jerusalem 91120, Israel; (N.R.); (F.-E.A.M.)
| |
Collapse
|
11
|
Zhang R, Qu J. The Mechanisms and Efficacy of Photobiomodulation Therapy for Arthritis: A Comprehensive Review. Int J Mol Sci 2023; 24:14293. [PMID: 37762594 PMCID: PMC10531845 DOI: 10.3390/ijms241814293] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) have a significant impact on the quality of life of patients around the world, causing significant pain and disability. Furthermore, the drugs used to treat these conditions frequently have side effects that add to the patient's burden. Photobiomodulation (PBM) has emerged as a promising treatment approach in recent years. PBM effectively reduces inflammation by utilizing near-infrared light emitted by lasers or LEDs. In contrast to photothermal effects, PBM causes a photobiological response in cells, which regulates their functional response to light and reduces inflammation. PBM's anti-inflammatory properties and beneficial effects in arthritis treatment have been reported in numerous studies, including animal experiments and clinical trials. PBM's effectiveness in arthritis treatment has been extensively researched in arthritis-specific cells. Despite the positive results of PBM treatment, questions about specific parameters such as wavelength, dose, power density, irradiation time, and treatment site remain. The goal of this comprehensive review is to systematically summarize the mechanisms of PBM in arthritis treatment, the development of animal arthritis models, and the anti-inflammatory and joint function recovery effects seen in these models. The review also goes over the evaluation methods used in clinical trials. Overall, this review provides valuable insights for researchers investigating PBM treatment for arthritis, providing important references for parameters, model techniques, and evaluation methods in future studies.
Collapse
Affiliation(s)
| | - Junle Qu
- Center for Biomedical Optics and Photonics and College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China;
| |
Collapse
|
12
|
Zarychta E, Bielawski K, Wrzeszcz K, Rhone P, Ruszkowska-Ciastek B. Unraveling the Angiogenic Puzzle: Pre-Treatment sVEGFR1 and sVEGFR2 Levels as Promising Prognostic Indicators in Early-Stage Breast Cancer Patients. Int J Mol Sci 2023; 24:13508. [PMID: 37686312 PMCID: PMC10487545 DOI: 10.3390/ijms241713508] [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: 08/01/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
Despite the advancements in breast cancer (BrC) diagnosis and treatment, a considerable proportion of patients with early-stage disease still experience local recurrence or metastasis. This study aimed to assess the levels of specific angiogenic parameters in the EDTA plasma of BrC patients before and after treatment and to explore their clinical and prognostic significance. The levels of vascular endothelial growth factor A (VEGF-A), soluble form of vascular endothelial growth factor receptor type 1 (sVEGFR1), and soluble form of vascular endothelial growth factor receptor type 2 (sVEGFR2) were measured in 84 early BrC patients, both prior to surgery and within a median time of nine months post-treatment. Prognostic significance was evaluated using Kaplan-Meier survival and Cox regression analyses. Linear regression models were employed to examine the independent impact of selected angiogenic factors on DFS in breast cancer patients. The results of uni- and multivariate analyses indicated that a pre-treatment concentration of sVEGFR1 above 30.99 pg/mL was associated with improved disease-free survival (DFS) (p < 0.0001 for both analyses), while a pre-treatment concentration of sVEGFR2 above 9475.67 pg/mL was associated with an increased risk of BrC relapse (p < 0.0001 for both analyses). Additionally, a post-treatment concentration of sVEGFR2 above 7361.71 pg/mL was associated with better overall survival (OS) based on the Kaplan-Meier survival analysis (p = 0.0141). Furthermore, linear regression models revealed a significant inverse association between pre-treatment levels of sVEGFR1 and the risk of relapse (standardized β -0.2578, p = 0.0499) and a significant positive association of VEGF-A levels with the risk of recurrence (standardized β 0.2958, p = 0.0308). In conclusion, the findings suggest that both pre- and post-treatment levels of sVEGFR1 and sVEGFR2 may hold promise as potential prognostic markers for BrC patients.
Collapse
Affiliation(s)
- Elżbieta Zarychta
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 9 Curie Sklodowska Street, 85-094 Bydgoszcz, Poland; (K.B.); (K.W.); (B.R.-C.)
- Invicta Fertility Clinic, 6 Zlota Street, 00-019 Warsaw, Poland
| | - Kornel Bielawski
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 9 Curie Sklodowska Street, 85-094 Bydgoszcz, Poland; (K.B.); (K.W.); (B.R.-C.)
| | - Katarzyna Wrzeszcz
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 9 Curie Sklodowska Street, 85-094 Bydgoszcz, Poland; (K.B.); (K.W.); (B.R.-C.)
| | - Piotr Rhone
- Clinical Ward of Breast Cancer and Reconstructive Surgery, Oncology Centre Prof. F. Łukaszczyk Memorial Hospital, 2 Romanowska Street, 85-796 Bydgoszcz, Poland;
| | - Barbara Ruszkowska-Ciastek
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 9 Curie Sklodowska Street, 85-094 Bydgoszcz, Poland; (K.B.); (K.W.); (B.R.-C.)
| |
Collapse
|
13
|
Ferre-Torres J, Noguera-Monteagudo A, Lopez-Canosa A, Romero-Arias JR, Barrio R, Castaño O, Hernandez-Machado A. Modelling of chemotactic sprouting endothelial cells through an extracellular matrix. Front Bioeng Biotechnol 2023; 11:1145550. [PMID: 37362221 PMCID: PMC10285466 DOI: 10.3389/fbioe.2023.1145550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Sprouting angiogenesis is a core biological process critical to vascular development. Its accurate simulation, relevant to multiple facets of human health, is of broad, interdisciplinary appeal. This study presents an in-silico model replicating a microfluidic assay where endothelial cells sprout into a biomimetic extracellular matrix, specifically, a large-pore, low-concentration fibrin-based porous hydrogel, influenced by chemotactic factors. We introduce a novel approach by incorporating the extracellular matrix and chemotactic factor effects into a unified term using a single parameter, primarily focusing on modelling sprouting dynamics and morphology. This continuous model naturally describes chemotactic-induced sprouting with no need for additional rules. In addition, we extended our base model to account for matrix sensing and degradation, crucial aspects of angiogenesis. We validate our model via a hybrid in-silico experimental method, comparing the model predictions with experimental results derived from the microfluidic setup. Our results underscore the intricate relationship between the extracellular matrix structure and angiogenic sprouting, proposing a promising method for predicting the influence of the extracellular matrix on angiogenesis.
Collapse
Affiliation(s)
- Josep Ferre-Torres
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | | | - Adrian Lopez-Canosa
- Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Spain
| | - J Roberto Romero-Arias
- Institute for Research in Applied Mathematics and Systems, National Autonomous University of Mexico , Mexico City, Mexico
| | - Rafael Barrio
- Institute of Physics, National Autonomous University of Mexico, Mexico City, Mexico
| | - Oscar Castaño
- Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona (UB), Barcelona, Spain
| | - Aurora Hernandez-Machado
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona (UB), Barcelona, Spain
| |
Collapse
|
14
|
Kou HS, Lo ST, Wang CC. One Single Tube Reaction of Aptasensor-Based Magnetic Sensing System for Selective Fluorescent Detection of VEGF in Plasma. BIOSENSORS 2023; 13:574. [PMID: 37366939 DOI: 10.3390/bios13060574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023]
Abstract
In this study, a simple, easy and convenient fluorescent sensing system for the detection of the vascular endothelial growth factor (VEGF) based on VEGF aptamers, aptamer-complementary fluorescence-labeled probe and streptavidin magnetic beads was developed in one single tube. The VEGF is the most important biomarker in cancer, and it is investigated that the serum VEGF level varied according to the different types and courses of cancers. Hence, efficient quantification of VEGF is able to improve the accuracy of cancer diagnoses and the precision of disease surveillance. In this research, the VEGF aptamer was designed to be able to bind with the VEGF by forming G-quadruplex secondary structures; then, the magnetic beads would capture the non-binding aptamers due to non-steric interference; and finally, the fluorescence-labeled probes were hybridized with the aptamers captured by the magnetic beads. Therefore, the fluorescent intensity in the supernatant would specifically reflect the present VEGF. After an overall optimization, the optimal conditions for the detection of VEGF were as followed, KCl, 50 μM; pH 7.0; aptamer, 0.1 μM; and magnetic beads, 10 μL (4 μg/μL). The VEGF could be well quantified within a range of 0.2-2.0 ng/mL in plasma, and the calibration curve possessed a good linearity (y = 1.0391x + 0.5471, r = 0.998). The detection limit (LOD) was calculated to be 0.0445 ng/mL according to the formula (LOD = 3.3 × σ/S). The specificity of this method was also investigated under the appearance of many other serum proteins, and the data showed good specificity in this aptasensor-based magnetic sensing system. This strategy provided a simple, sensitive and selective biosensing platform for the detection of serum VEGF. Finally, it was expected that this detection technique can be used to promote more clinical applications.
Collapse
Affiliation(s)
- Hwang-Shang Kou
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shao-Tsung Lo
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chun-Chi Wang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
15
|
Duan R, Pan H, Li D, Liao S, Han B. Ergothioneine improves myocardial remodeling and heart function after acute myocardial infarction via S-glutathionylation through the NF-ĸB dependent Wnt5a-sFlt-1 pathway. Eur J Pharmacol 2023; 950:175759. [PMID: 37121564 DOI: 10.1016/j.ejphar.2023.175759] [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: 02/09/2022] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/02/2023]
Abstract
Myocardial infarction (MI) remains the leading cause of cardiovascular death worldwide. Studies have shown that soluble fms-like tyrosine kinase-1 (sFlt-1) has a harmful effect on the heart after MI. However, ergothioneine (ERG) has been shown to have protective effects in rats with preeclampsia by reducing circulating levels of sFlt-1. In this study, we aimed to investigate the mechanism by which ERG protects the heart after MI in rats. Our results indicate that treatment with 10 mg/kg ERG for 7 days can improve cardiac function as determined by echocardiography. Additionally, ERG can reduce the size of the damaged area, prevent heart remodeling, fibrosis, and reduce cardiomyocyte death after MI. To explain the mechanism behind the cardioprotective effects of ERG, we conducted several experiments. We observed a significant reduction in the expression of monocyte chemoattractant protein-1 (MCP-1), p65, and p-p65 proteins in heart tissues of ERG-treated rats compared to the control group. ELISA results also showed that ERG significantly reduced plasma levels of sFlt-1. Using Glutaredoxin-1 (GLRX) and CD31 immunofluorescence, we found that GLRX was expressed in clusters in the myocardial tissue surrounding the coronary artery, and ERG can reduce the expression of GLRX caused by MI. In vitro experiments using a human coronary artery endothelial cell (HCAEC) hypoxia model confirmed that ERG can reduce the expression of sFlt-1, GLRX, and Wnt5a. These findings suggest that ERG protects the heart from MI damage by reducing s-glutathionylation through the NF-ĸB-dependent Wnt5a-sFlt-1 pathway.
Collapse
Affiliation(s)
- Rui Duan
- Department of Cardiology, Xuzhou Central Hospital, Jiangsu, PR China
| | - Haotian Pan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, PR China
| | - DongCheng Li
- Department of Cardiology, Huai'an First People's Hospital, Jiangsu, PR China
| | - Shengen Liao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, PR China.
| | - Bing Han
- Department of Cardiology, Xuzhou Central Hospital, Jiangsu, PR China.
| |
Collapse
|
16
|
Huang X, Jia L, Jia Y, Xu X, Wang R, Wei M, Li H, Peng H, Wei Y, He Q, Wang K. sFlt-1-enriched exosomes induced endothelial cell dysfunction and a preeclampsia-like phenotype in mice. Cytokine 2023; 166:156190. [PMID: 37062152 DOI: 10.1016/j.cyto.2023.156190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/18/2023]
Abstract
Preeclampsia (PE) is a hypertensive disorder of pregnancy characterized by maternal endothelial dysfunction and end-organ damage. Our previous work demonstrated that PE patient-derived exosomes contained higher levels of soluble FMS-like tyrosine kinase-1 (sFlt-1) and significantly induced endothelial dysfunction and PE development. However, the mechanisms underlying the effect of sFlt-1-enriched exosomes (sFlt-1-Exo) on PE development are poorly characterized. Here, we revealed that trophoblast-derived sFlt-1-Exo treatment induced significant inhibition of human umbilical vein endothelial cell (HUVEC) migration and tube formation, as well as an increase in sFlt-1 secretion. Mechanistically, we found that the increased sFlt-1 secretion in the cell culture medium was attributed to enhanced transcription of sFlt-1 in HUVECs. Importantly, we observed that treating pregnant mice with sFlt-1-Exo or recombinant mouse sFlt-1 triggered a preeclampsia-like phenotype, characterized by elevated blood pressure, proteinuria, increased plasma sFlt-1 and adverse pregnancy outcomes. These results strongly suggested that sFlt-1-Exo-induced endothelial dysfunction could be partially attributed to the upregulation of sFlt-1 in endothelial cells, potentially leading to the development of a preeclampsia-like phenotype in mice.
Collapse
Affiliation(s)
- Xiaojie Huang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Linyan Jia
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanhui Jia
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xianghong Xu
- Department of Biobank, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ruixue Wang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mengtian Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Han Li
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Peng
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yingying Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qizhi He
- Department of Pathology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
17
|
Zhou H, Wang Z, Dong Y, Alhaskawi A, Tu T, Hasan Abdullah Ezzi S, Goutham Kota V, Hasan Abdulla Hasan Abdulla M, Li P, Wu B, Chen Y, Lu H. New advances in treatment of skin malignant tumors with nanosecond pulsed electric field: A literature review. Bioelectrochemistry 2023; 150:108366. [PMID: 36641842 DOI: 10.1016/j.bioelechem.2023.108366] [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: 08/21/2022] [Revised: 12/05/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
BACKGROUND Nanosecond pulsed electric field, with its unique bioelectric effect, has shown broad application potential in the field of tumor therapy, especially in malignant tumors and skin tumors. MAIN BODY In this paper, we discuss the therapeutic effects and mechanisms of nanosecond pulsed electric field on three common skin cancers, namely, malignant melanoma, squamous cell carcinoma and basal cell carcinoma, as well as its application to other benign skin diseases and future development and improvement directions. CONCLUSION In general, nanosecond pulsed electric field mainly exerts its ablative effect on tumors through subcellular membrane electroporation effect. It is cell type-specific, has less thermal damage, and can have synergistic effect with chemotherapy drugs, making it a very promising new method for tumor treatment.
Collapse
Affiliation(s)
- Haiying Zhou
- Department of Orthopedics, The First Affiliated Hospital, College of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Zewei Wang
- Zhejiang University School of Medicine, #866 Yuhangtang Road, Hangzhou, Zhejiang Province 310058, PR China
| | - Yanzhao Dong
- Department of Orthopedics, The First Affiliated Hospital, College of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Ahmad Alhaskawi
- Department of Orthopedics, The First Affiliated Hospital, College of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Tian Tu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | | | - Vishnu Goutham Kota
- Zhejiang University School of Medicine, #866 Yuhangtang Road, Hangzhou, Zhejiang Province 310058, PR China
| | | | - Pengfei Li
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Bin Wu
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Ruidi Biotech Ltd. #2959 Yuhangtang Road, Hangzhou, Zhejiang Province 310000, PR China
| | - Yonggang Chen
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Ruidi Biotech Ltd. #2959 Yuhangtang Road, Hangzhou, Zhejiang Province 310000, PR China
| | - Hui Lu
- Department of Orthopedics, The First Affiliated Hospital, College of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, #866 Yuhangtang Road, Hangzhou, Zhejiang Province 310058, PR China.
| |
Collapse
|
18
|
Mi P, Liu JL, Qi BP, Wei BM, Xu CZ, Zhu L. Stem cell-derived exosomes for chronic wound repair. Cell Tissue Res 2023; 391:419-423. [PMID: 36705748 DOI: 10.1007/s00441-023-03742-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/18/2023] [Indexed: 01/28/2023]
Abstract
Stem cells possess the capability of self-renewal and multipotency, which endows them with great application potential in wound repair fields. Yet, several problems including immune concerns, ethical debates, and oncogenicity impede the broad and deep advance of stem cell-based products. Recently, owing to their abundant resources, excellent biocompatibility, and ease of being engineered, stem cell-derived exosomes were proved to be promising nanomedicine for curing chronic wounds. What is more, stem cell-derived exosomes are almost the mini record of their maternal cells, which even equipped them with the unique characteristics of stem cells. Chronic wound healing efficacy is dominated by several complicated factors, especially the excessive inflammation conditions and impaired vessels. Therefore, this review tries to concentrate on the current advances of stem cell-derived exosomes for reducing inflammation and promoting angiogenesis in chronic wound healing processes. Last but not least, the existing limitations and future perspectives of stem cell-derived exosomes for chronic wound treatment are also outlined.
Collapse
Affiliation(s)
- Peng Mi
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Changqing Garden, Wuhan, 430023, Hubei, China
| | - Jia-Lin Liu
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Changqing Garden, Wuhan, 430023, Hubei, China
| | - Bao-Ping Qi
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Enshi, 445000, Hubei, China. .,School of Chemistry and Environmental Engineering, Hubei Minzu University, Xueyuan Road, Enshi, 445000, Hubei, China.
| | - Ben-Mei Wei
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Changqing Garden, Wuhan, 430023, Hubei, China
| | - Cheng-Zhi Xu
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Changqing Garden, Wuhan, 430023, Hubei, China
| | - Lian Zhu
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Changqing Garden, Wuhan, 430023, Hubei, China.
| |
Collapse
|
19
|
Adini A, Ko VH, Puder M, Louie SM, Kim CF, Baron J, Matthews BD. PR1P, a VEGF-stabilizing peptide, reduces injury and inflammation in acute lung injury and ulcerative colitis animal models. Front Immunol 2023; 14:1168676. [PMID: 37187742 PMCID: PMC10175756 DOI: 10.3389/fimmu.2023.1168676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) and Ulcerative Colitis (UC) are each characterized by tissue damage and uncontrolled inflammation. Neutrophils and other inflammatory cells play a primary role in disease progression by acutely responding to direct and indirect insults to tissue injury and by promoting inflammation through secretion of inflammatory cytokines and proteases. Vascular Endothelial Growth Factor (VEGF) is a ubiquitous signaling molecule that plays a key role in maintaining and promoting cell and tissue health, and is dysregulated in both ARDS and UC. Recent evidence suggests a role for VEGF in mediating inflammation, however, the molecular mechanism by which this occurs is not well understood. We recently showed that PR1P, a 12-amino acid peptide that binds to and upregulates VEGF, stabilizes VEGF from degradation by inflammatory proteases such as elastase and plasmin thereby limiting the production of VEGF degradation products (fragmented VEGF (fVEGF)). Here we show that fVEGF is a neutrophil chemoattractant in vitro and that PR1P can be used to reduce neutrophil migration in vitro by preventing the production of fVEGF during VEGF proteolysis. In addition, inhaled PR1P reduced neutrophil migration into airways following injury in three separate murine acute lung injury models including from lipopolysaccharide (LPS), bleomycin and acid. Reduced presence of neutrophils in the airways was associated with decreased pro-inflammatory cytokines (including TNF-α, IL-1β, IL-6) and Myeloperoxidase (MPO) in broncho-alveolar lavage fluid (BALF). Finally, PR1P prevented weight loss and tissue injury and reduced plasma levels of key inflammatory cytokines IL-1β and IL-6 in a rat TNBS-induced colitis model. Taken together, our data demonstrate that VEGF and fVEGF may each play separate and pivotal roles in mediating inflammation in ARDS and UC, and that PR1P, by preventing proteolytic degradation of VEGF and the production of fVEGF may represent a novel therapeutic approach to preserve VEGF signaling and inhibit inflammation in acute and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Avner Adini
- Vascular Biology Program, Children’s Hospital Boston and Harvard Medical School, Boston, MA, United States
- Department of Medicine, Boston Children’s Hospital, Boston, MA, United States
- *Correspondence: Avner Adini,
| | - Victoria H. Ko
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States
| | - Mark Puder
- Department of Surgery, Boston Children’s Hospital, Boston, MA, United States
| | - Sharon M. Louie
- Stem Cell Program and Divisions of Hematology/Oncology, Boston Children’s Hospital, Boston, MA, United States
| | - Carla F. Kim
- Stem Cell Program and Divisions of Hematology/Oncology, Boston Children’s Hospital, Boston, MA, United States
| | - Joseph Baron
- Janus Biotherapeutics, Inc, Wellesley, MA, United States
| | - Benjamin D. Matthews
- Vascular Biology Program, Children’s Hospital Boston and Harvard Medical School, Boston, MA, United States
- Department of Medicine, Boston Children’s Hospital, Boston, MA, United States
| |
Collapse
|
20
|
β-carotene alleviates LPS-induced inflammation through regulating STIM1/ORAI1 expression in bovine mammary epithelial cells. Int Immunopharmacol 2022; 113:109377. [DOI: 10.1016/j.intimp.2022.109377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/12/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
|
21
|
Xue S, Wang L, Cai J. Sulfono-γ-AApeptides as Protein Helical Domain Mimetics to Manipulate the Angiogenesis. Chembiochem 2022; 23:e202200298. [PMID: 36006398 PMCID: PMC9741949 DOI: 10.1002/cbic.202200298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/23/2022] [Indexed: 02/03/2023]
Abstract
Sulfono-γ-AApeptides recently developed in our group have been proven to be a new class of unnatural foldamer with well-defined helical structure and have been demonstrated to mimic protein helical domains and disrupt biomedically relevant protein-protein interactions (PPIs). Based on our design concept in a recent report, we discovered two similar sulfono-γ-AApeptides V2 and V3 which were designed to mimic the VEGF N-terminal helix α1 known to directly interact with VEGFRs. Interestingly, V2 was shown to possess the pro-angiogenic effect, whereas V3 was proved to be a potent inhibitor for angiogenesis. We speculate that the distinct angiogenesis signaling was due to the selective binding of the two molecules to VEGFR1 and VEGFR2, respectively. Together with their remarkable resistance to proteolytic degradation, relatively small sizes, and amenability to modification with diverse functional groups, V2 and V3 could serve as lead molecules for the development of potential therapeutic agents and molecular probes. These findings highlight sulfono-γ-AApeptides as an alternative paradigm to mimic the α-helical domain to modulate a wide variety of PPIs in the future.
Collapse
Affiliation(s)
- Songyi Xue
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, USA
| | - Lei Wang
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, USA
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, USA
| |
Collapse
|
22
|
Zhao P, Zhang N, An Z. Engineering antibody and protein therapeutics to cross the blood-brain barrier. Antib Ther 2022; 5:311-331. [PMID: 36540309 PMCID: PMC9759110 DOI: 10.1093/abt/tbac028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/10/2022] [Accepted: 11/01/2022] [Indexed: 08/17/2023] Open
Abstract
Diseases in the central nervous system (CNS) are often difficult to treat. Antibody- and protein-based therapeutics hold huge promises in CNS disease treatment. However, proteins are restricted from entering the CNS by the blood-brain barrier (BBB). To achieve enhanced BBB crossing, antibody-based carriers have been developed by utilizing the endogenous macromolecule transportation pathway, known as receptor-mediated transcytosis. In this report, we first provided an overall review on key CNS diseases and the most promising antibody- or protein-based therapeutics approved or in clinical trials. We then reviewed the platforms that are being explored to increase the macromolecule brain entry to combat CNS diseases. Finally, we have analyzed the lessons learned from past experiences and have provided a perspective on the future engineering of novel delivery vehicles for antibody- and protein-based therapies for CNS diseases.
Collapse
Affiliation(s)
- Peng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| |
Collapse
|
23
|
Goswami AG, Basu S, Huda F, Pant J, Ghosh Kar A, Banerjee T, Shukla VK. An appraisal of vascular endothelial growth factor (VEGF): the dynamic molecule of wound healing and its current clinical applications. Growth Factors 2022; 40:73-88. [PMID: 35584274 DOI: 10.1080/08977194.2022.2074843] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Angiogenesis is a critical step of wound healing, and its failure leads to chronic wounds. The idea of restoring blood flow to the damaged tissues by promoting neo-angiogenesis is lucrative and has been researched extensively. Vascular endothelial growth factor (VEGF), a key dynamic molecule of angiogenesis has been investigated for its functions. In this review, we aim to appraise its biology, the comprehensive role of this dynamic molecule in the wound healing process, and how this knowledge has been translated in clinical application in various types of wounds. Although, most laboratory research on the use of VEGF is promising, its clinical applications have not met great expectations. We discuss various lacunae that might exist in making its clinical application unsuccessful for commercial use, and provide insight to the foundation for future research.
Collapse
Affiliation(s)
- Aakansha Giri Goswami
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Somprakas Basu
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Farhanul Huda
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Jayanti Pant
- Department of Physiology, All India Institute of Medical Sciences, Rishikesh, India
| | - Amrita Ghosh Kar
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Tuhina Banerjee
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vijay Kumar Shukla
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
24
|
Characterization of the Expression of Angiogenic Factors in Cutaneous Squamous Cell Carcinoma of Domestic Cats. Vet Sci 2022; 9:vetsci9070375. [PMID: 35878392 PMCID: PMC9351683 DOI: 10.3390/vetsci9070375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is a common malignant skin cancer with a significant impact on health, and it is important to determine the degree of reliance of CSCC on angiogenesis for growth and metastasis. Major regulators of angiogenesis are the vascular endothelial growth factor (VEGF) family and their associated receptors. Alternative pre-mRNA splicing produces multiple isoforms of VEGF-A and PLGF with distinct biological properties. Several studies highlight the function of VEGF-A in CSCC, but there are no studies of the different isoforms of VEGF-A and PLGF for this neoplasm. We characterized the expression of three isoforms of VEGF-A, two isoforms of PLGF, and their receptors in cat CSCC biopsies compared to normal haired skin (NHS). Although our results revealed no significant changes in transcript levels of panVEGF-A or their isoforms, the mRNA levels of PLGF I and the receptors Flt-1 and KDR were downregulated in CSCC compared to NHS. Differences were observed in ligand:receptor mRNA expression ratio, with the expression of VEGF-A relative to its receptor KDR higher in CSCC, which is consistent with our hypothesis and prior human SCC studies. Immunolocalization in tissue showed increased expression of all measured factors and receptors in tumor cells compared to NHS and surrounding vasculature. We conclude that the factors measured may play a pivotal role in CSCC growth, although further studies are needed to clarify the role of angiogenic factors in feline CSCC.
Collapse
|
25
|
Luo G, Jia Y, Hu Y, Wu F, Wang M, Chen X. Practical synthesis of ECH and epoxyquinols A and B from (-)-shikimic acid. Org Biomol Chem 2022; 20:4608-4615. [PMID: 35608102 DOI: 10.1039/d2ob00559j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An efficient synthesis of ECH, epoxyquinols A and B, and two bioactive analogs EqM and RKTS-33 has been completed starting from (-)-shikimic acid. Rapid establishment of the desired epoxyquinol core is facilitated through a key allylic oxidation with high stereoselectivity, which is achieved by fine tuning the cyclohexene substrate structure and reaction conditions.
Collapse
Affiliation(s)
- Guiyin Luo
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, PR China.
| | - Yuanliang Jia
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, PR China.
| | - Yue Hu
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, PR China.
| | - Folei Wu
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, PR China.
| | - Maolin Wang
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, PR China.
| | - Xiaochuan Chen
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, PR China.
| |
Collapse
|
26
|
Vascular mimicry: A potential therapeutic target in breast cancer. Pathol Res Pract 2022; 234:153922. [DOI: 10.1016/j.prp.2022.153922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/19/2022] [Accepted: 04/24/2022] [Indexed: 10/18/2022]
|
27
|
Liu Y, Zhang Q, Gao X, Wang T. Study on lipid nanomicelles targeting placenta for the treatment of preeclampsia. J Drug Target 2022; 30:894-909. [PMID: 35502921 DOI: 10.1080/1061186x.2022.2068558] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In view of the serious clinical harm of preeclampsia and the lack of effective treatment methods, a PEG-modified lipid hybrid micelle was designed with folic acid molecule on the surface, containing siRNA, targeted delivery to the placenta, interfering the expression of sFlt-1 and treating preeclampsia. In this paper, the preparation and characterization of lipid hybrid micelles were investigated in detail, the cytology in vitro and in vivo distribution, pharmacodynamics, safety and action mechanism of the preparation were studied, which laid a foundation for gene therapy of preeclampsia.
Collapse
Affiliation(s)
- Yang Liu
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| | - Qimeng Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| | - Xingli Gao
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| | - Tong Wang
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| |
Collapse
|
28
|
Vore AS, Barney TM, Deak MM, Varlinskaya EI, Deak T. Adolescent intermittent ethanol exposure produces Sex-Specific changes in BBB Permeability: A potential role for VEGFA. Brain Behav Immun 2022; 102:209-223. [PMID: 35245677 PMCID: PMC9277567 DOI: 10.1016/j.bbi.2022.02.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/31/2022] [Accepted: 02/27/2022] [Indexed: 12/20/2022] Open
Abstract
Binge drinking that typically begins during adolescence can have long-lasting neurobehavioral consequences, including alterations in the central and peripheral immune systems. Central and peripheral inflammation disrupts blood-brain barrier (BBB) integrity and exacerbates pathology in diseases commonly associated with disturbed BBB function. Thus, the goal of the present studies was to determine long-lasting effects of adolescent intermittent ethanol (AIE) on BBB integrity. For AIE, male and female Sprague Dawley rats were repeatedly exposed to ethanol (4 g/kg, intragastrically) or water during adolescence between postnatal day (P) 30 and P50. In adulthood (∼P75), rats were challenged with fluorescein isothiocyanate (FITC)-tagged Dextran of varying molecular weights (4, 20, & 70 kDa) for assessment of BBB permeability using gross tissue fluorometry (Experiment 1). Experiment 2 extended these effects using immunofluorescence, adding an adult ethanol-exposed group to test for a specific developmental vulnerability. Finally, as a first test of hypothesized mechanism, Experiment 3 examined the effect of AIE on Vascular Endothelial Growth Factor A (VEGFA) and its co-localization with pericytes (identified through expression of platelet derived growth factor receptor beta (PDGFRβ), a key regulatory cell embedded within the BBB. Male, but not female, rats with a history of AIE showed significantly increased dextran permeability in the nucleus accumbens (NAc), cingulate prefrontal cortex (cPFC), and amygdala (AMG). Similar increases in dextran were observed in the hippocampus (HPC) and ventral tegmental area (VTA) of male rats with a history of AIE or equivalent ethanol exposure during adulthood. No changes in BBB permeability were evident in females. When VEGFa expression was examined, male rats exposed to AIE were challenged with 3.5 g/kg ethanol (i.p.) or vehicle acutely in adulthood to assess long-lasting versus acute actions of ethanol. Adult rats with a history of AIE showed significantly fewer total cells expressing VEGFa in the AMG and dHPC following the acute ethanol challenge in adulthood. They also showed a significant reduction in the number of PDGFRβ positive cells that also expressed VEGFa signal. The anatomical distribution of these effects corresponded with increased BBB permeability after AIE (i.e., differential effects in the PVN, AMG, and dHPC). These studies demonstrated sex-specific effects of AIE, with males, but not females, demonstrating long-term increases in BBB permeability that correlated with changes in VEGFa and PDGFRβ protein, two factors known to influence BBB permeability.
Collapse
Affiliation(s)
| | | | | | | | - Terrence Deak
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton, NY 13902-6000.
| |
Collapse
|
29
|
Liu X, Li Y, Zhang Q, Pan Q, Zheng P, Dai X, Bai Z, Zhu W. Design, Synthesis, and Biological Evaluation of [1,2,4]triazolo[4,3-a] Pyrazine Derivatives as Novel Dual c-Met/VEGFR-2 Inhibitors. Front Chem 2022; 10:815534. [PMID: 35464202 PMCID: PMC9019572 DOI: 10.3389/fchem.2022.815534] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
In this study, we designed and synthesized a series of novel [1,2,4]triazolo [4,3-a]pyrazine derivatives, and evaluated them for their inhibitory activities toward c-Met/VEGFR-2 kinases and antiproliferative activities against tested three cell lines in vitro. Most of the compounds showed satisfactory activity compared with lead compound foretinib. Among them, the most promising compound 17l exhibited excellent antiproliferative activities against A549, MCF-7, and Hela cancer cell lines with IC50 values of 0.98 ± 0.08, 1.05 ± 0.17, and 1.28 ± 0.25 µM, respectively, as well as excellent kinase inhibitory activities (c-Met IC50 = 26.00 nM and VEGFR-2 IC50 = 2.6 µM). Moreover, compound 17l inhibited the growth of A549 cells in G0/G1 phase in a dose-dependent manner, and induced the late apoptosis of A549 cells. Its intervention on intracellular c-Met signaling of A549 was verified by the result of Western blot. Fluorescence quantitative PCR showed that compound 17l inhibited the growth of A549 cells by inhibiting the expression of c-Met and VEGFR-2, and its hemolytic toxicity was low. Molecular docking and molecular dynamics simulation indicated that compound 17l could bind to c-Met and VEGFR-2 protein, which was similar to that of foretinib.
Collapse
Affiliation(s)
- Xiaobo Liu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, China
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Yuzhen Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Qian Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Qingshan Pan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Xinyang Dai
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Zhaoshi Bai
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zhaoshi Bai, ; Wufu Zhu,
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang, China
- *Correspondence: Zhaoshi Bai, ; Wufu Zhu,
| |
Collapse
|
30
|
Li X, Li Y, Wang Y, Liu F, Liu Y, Liang J, Zhan R, Wu Y, Ren H, Zhang X, Liu J. Sinensetin suppresses angiogenesis in liver cancer by targeting the VEGF/VEGFR2/AKT signaling pathway. Exp Ther Med 2022; 23:360. [PMID: 35493423 PMCID: PMC9019764 DOI: 10.3892/etm.2022.11287] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/15/2022] [Indexed: 11/06/2022] Open
Abstract
Sinensetin (SIN) is a polymethoxy flavone primarily present in citrus fruits. This compound has demonstrated anticancer activity. However, the underlying mechanism of its action has not been fully understood. The present study investigated the impact of SIN on angiogenesis in a liver cancer model. In a murine xenograft tumor model, SIN inhibited the growth of HepG2/C3A human liver hepatoma cell-derived tumors and reduced the expression levels of platelet/endothelial cell adhesion molecule-1 and VEGF. In HepG2/C3A cells, SIN repressed VEGF expression by downregulating hypoxia-inducible factor expression. In cultured human umbilical vein endothelial cells, SIN increased apoptosis and repressed migration and tube formation. In addition, SIN decreased the phosphorylation of VEGFR2 and inhibited the AKT signaling pathway. Molecular docking demonstrated that the VEGFR2 core domain effectively combined with SIN at various important residues. Collectively, these data suggested that SIN inhibited liver cancer angiogenesis by regulating VEGF/VEGFR2/AKT signaling.
Collapse
Affiliation(s)
- Xiaο Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong 250355, P.R. China
| | - Yan Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong 250355, P.R. China
| | - Yuan Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong 250355, P.R. China
| | - Fuhong Liu
- Laboratory 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
| | - Yanjun Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong 250355, P.R. China
| | - Jiangjiu Liang
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, P.R. China
| | - Rucai Zhan
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, P.R. China
| | - Yue Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong 250355, P.R. China
| | - He Ren
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong 250355, P.R. China
| | - Xiuyuan Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong 250355, P.R. China
| | - Ju Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong 250355, P.R. China
| |
Collapse
|
31
|
Wang A, Madden LA, Paunov VN. Vascularized Co-Culture Clusteroids of Primary Endothelial and Hep-G2 Cells Based on Aqueous Two-Phase Pickering Emulsions. Bioengineering (Basel) 2022; 9:bioengineering9030126. [PMID: 35324815 PMCID: PMC8945860 DOI: 10.3390/bioengineering9030126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/05/2022] [Accepted: 03/09/2022] [Indexed: 11/16/2022] Open
Abstract
Three-dimensional cell culture has been extensively involved in biomedical applications due to its high availability and relatively mature biochemical properties. However, single 3D cell culture models based on hydrogel or various scaffolds do not meet the more in-depth requirements of in vitro models. The necrotic core formation inhibits the utilization of the 3D cell culture ex vivo as oxygen permeation is impaired in the absence of blood vessels. We report a simple method to facilitate the formation of angiogenic HUVEC (human umbilical vein endothelial cells) and Hep-G2 (hepatocyte carcinoma model) co-culture 3D clusteroids in a water-in-water (w/w) Pickering emulsions template which can overcome this limitation. This method enabled us to manipulate the cells proportion in order to achieve the optimal condition for stimulating the production of various angiogenic protein markers in the co-cultured clusteroids. The HUVEC cells respond to the presence of Hep-G2 cells and their byproducts by forming endothelial cell sprouts in Matrigel without the exogenous addition of vascular endothelial growth factor (VEGF) or other angiogenesis inducers. This culture method can be easily replicated to produce other types of cell co-culture spheroids. The w/w Pickering emulsion template can facilitate the fabrication of 3D co-culture models to a great extent and be further utilized in drug testing and tissue engineering applications.
Collapse
Affiliation(s)
- Anheng Wang
- Department of Chemistry, University of Hull, Hull HU6 7RX, UK;
| | - Leigh A. Madden
- Department of Biomedical Sciences, University of Hull, Hull HU6 7RX, UK;
| | - Vesselin N. Paunov
- Department of Chemistry, Nazarbayev University, Nur-Sultan 010000, Kazakhstan
- Correspondence:
| |
Collapse
|
32
|
Negi R, Haritha V, Aziz N, Siddiqui AH. Biochemical markers in the pathogenesis of preeclampsia: novel link between placental growth factor and interleukin-6. Mol Cell Biochem 2022; 477:1765-1774. [PMID: 35292877 DOI: 10.1007/s11010-022-04403-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 02/25/2022] [Indexed: 11/29/2022]
Abstract
Preeclampsia (PE) is a multisystem disorder of pregnancy characterized by sudden onset of hypertension and proteinuria. The appearance and diagnosis of the disease remain elusive and the only treatment is the termination of pregnancy. The onset of the disease is preceded by a shift in the balance of the angiogenic and antiangiogenic proteins in the maternal circulation. We surmised that the assessment of the levels of these proteins during pregnancy could lead to a proper diagnosis of the disease. In this study, we determined the levels of angiogenic and antiangiogenic proteins and IL-6 in maternal circulation during normotensive and hypertensive pregnancy, including PE. Serum isolated from pregnant women during antenatal visits was used to determine the concentrations of these proteins. The levels of antiangiogenic proteins, sFlt-1 and sEng, were higher in hypertensive disorders [gestational hypertension (GH), mild PE, and PE] of pregnancy and were significantly higher for PE than for GH. The levels of sFlt-1 and sEng were higher in PE samples compared to those in GH and NT samples. These proteins may have contributed to increased blood pressure. The levels of PlGF were decreased in pregnant women having GH, mild PE, and PE. The levels of the inflammatory intermediate, IL-6, were increased in PE samples compared to those in the GH and normotensive samples. The evaluation of the altered levels of antiangiogenic and angiogenic proteins can be useful for diagnosis of PE.
Collapse
Affiliation(s)
- Rahul Negi
- School of Medical Sciences, University of Hyderabad, Professor C. R. Rao Road, Gachibowli, Hyderabad, TS, 500046, India
| | - Vemanamanda Haritha
- Fernandez Hosp Educational & Research Foundation, Bogulkunta, Hyderabad, TS, 500001, India
| | - Nuzhat Aziz
- Fernandez Hosp Educational & Research Foundation, Bogulkunta, Hyderabad, TS, 500001, India
| | - Athar H Siddiqui
- School of Medical Sciences, University of Hyderabad, Professor C. R. Rao Road, Gachibowli, Hyderabad, TS, 500046, India.
| |
Collapse
|
33
|
Mauricio R, Singh K, Sanghavi M, Ayers CR, Rohatgi A, Vongpatanasin W, de Lemos JA, Khera A. Soluble Fms-like tyrosine kinase-1 (sFlt-1) is associated with subclinical and clinical ASCVD: The Dallas Heart Study. Atherosclerosis 2022; 346:46-52. [DOI: 10.1016/j.atherosclerosis.2022.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/20/2022] [Accepted: 02/25/2022] [Indexed: 11/02/2022]
|
34
|
Tosoc JPS, Nuñeza OM, Sudha T, Darwish NHE, Mousa SA. Anticancer Effects of the Corchorus olitorius Aqueous Extract and Its Bioactive Compounds on Human Cancer Cell Lines. Molecules 2021; 26:molecules26196033. [PMID: 34641577 PMCID: PMC8513029 DOI: 10.3390/molecules26196033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/25/2021] [Accepted: 09/29/2021] [Indexed: 11/18/2022] Open
Abstract
Corchorus olitorius is a common, leafy vegetable locally known as “Saluyot” in the Philippines. Several studies have reported on its various pharmacological properties, such as antioxidant, anti-inflammatory, analgesic, and anticancer properties. However, little is known about its effects on angiogenesis. This study aimed to evaluate the anticancer properties, such as the antiproliferative, anti-angiogenic, and antitumor activities, of the C. olitorius aqueous extract (CO) and its bioactive compounds, chlorogenic acid (CGA) and isoquercetin (IQ), against human melanoma (A-375), gastric cancer (AGS), and pancreatic cancer (SUIT-2), using in vitro and in ovo biological assays. The detection and quantification of CGA and IQ in CO were achieved using LC-MS/MS analysis. The antiproliferative, anti-angiogenic, and antitumor activities of CO, CGA, and IQ against A-375, AGS, and SUIT-2 cancer cell lines were evaluated using MTT and CAM assays. CGA and IQ were confirmed to be present in CO. CO, CGA, and IQ significantly inhibited the proliferation of A-375, AGS, and SUIT-2 cancer cells in a dose-dependent manner after 48 h of treatment. Tumor angiogenesis (hemoglobin levels) of A-375 and AGS tumors was significantly inhibited by CO, CGA, IQ, and a CGA–IQ combination. The growth of implanted A-375 and AGS tumors was significantly reduced by CO, CGA, IQ, and a CGA–IQ combination, as measured in tumor weight. Our investigation provides new evidence to show that CO has promising anticancer effects on various types of human cancer cells. CO and its compounds are potential nutraceutical products that could be used for cancer treatment.
Collapse
Affiliation(s)
- John Paul Sese Tosoc
- Department of Biological Sciences, College of Science and Mathematics, Mindanao State University-Iligan Institute of Technology, Iligan City 9200, Philippines;
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (N.H.E.D.); (S.A.M.)
- Correspondence: ; Tel.: +63-083-520-7969
| | - Olga Macas Nuñeza
- Department of Biological Sciences, College of Science and Mathematics, Mindanao State University-Iligan Institute of Technology, Iligan City 9200, Philippines;
| | - Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (N.H.E.D.); (S.A.M.)
| | - Noureldien H. E. Darwish
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (N.H.E.D.); (S.A.M.)
- Hematology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (N.H.E.D.); (S.A.M.)
| |
Collapse
|
35
|
Targeting Protein Kinase C in Glioblastoma Treatment. Biomedicines 2021; 9:biomedicines9040381. [PMID: 33916593 PMCID: PMC8067000 DOI: 10.3390/biomedicines9040381] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma (GBM) is the most frequent and aggressive primary brain tumor and is associated with a poor prognosis. Despite the use of combined treatment approaches, recurrence is almost inevitable and survival longer than 14 or 15 months after diagnosis is low. It is therefore necessary to identify new therapeutic targets to fight GBM progression and recurrence. Some publications have pointed out the role of glioma stem cells (GSCs) as the origin of GBM. These cells, with characteristics of neural stem cells (NSC) present in physiological neurogenic niches, have been proposed as being responsible for the high resistance of GBM to current treatments such as temozolomide (TMZ). The protein Kinase C (PKC) family members play an essential role in transducing signals related with cell cycle entrance, differentiation and apoptosis in NSC and participate in distinct signaling cascades that determine NSC and GSC dynamics. Thus, PKC could be a suitable druggable target to treat recurrent GBM. Clinical trials have tested the efficacy of PKCβ inhibitors, and preclinical studies have focused on other PKC isozymes. Here, we discuss the idea that other PKC isozymes may also be involved in GBM progression and that the development of a new generation of effective drugs should consider the balance between the activation of different PKC subtypes.
Collapse
|
36
|
Exosomes derived from macrophages upon cobalt ion stimulation promote angiogenesis. Colloids Surf B Biointerfaces 2021; 203:111742. [PMID: 33838581 DOI: 10.1016/j.colsurfb.2021.111742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/21/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
Angiogenesis is critical for tissue repair and regeneration, including implant osseointegration. It is well known that macrophages exert immunomodulatory functions in angiogenesis. However, whether macrophage-derived exosomes participate in the process is still unclear. Cobalt (Co) ions are frequently used as implant additives to mimic hypoxic microenvironment, which can induce angiogenesis through stabilizing hypoxia inducible factor-1α (HIF-1α) of macrophages and endothelial cells (ECs). The present work attempts to investigate whether exosomes derived from macrophages upon Co ion stimulation can mediate angiogenesis and the possible mechanism. The results show that the exosomes promote endothelial migration and angiogenesis in vitro and in vivo, particularly when Co ion concentration is 200 μM. Further studies reveal that the exosomes upregulating nitric oxide (NO), vascular endothelial growth factor (VEGF), and integrin β1 expression may be the underlying mechanism of the observations. Our findings provide new insights for Co ion mediated macrophage-EC communication and surface design of biomaterials from the perspective of pro-angiogenesis.
Collapse
|
37
|
Fonseca BM, Cunha SC, Gonçalves D, Mendes A, Braga J, Correia-da-Silva G, Teixeira NA. Decidual NK cell-derived conditioned medium from miscarriages affects endometrial stromal cell decidualisation: endocannabinoid anandamide and tumour necrosis factor-α crosstalk. Hum Reprod 2021; 35:265-274. [PMID: 31990346 DOI: 10.1093/humrep/dez260] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/04/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION What are the effects of endocannabinoid anandamide (AEA) in uterine natural killer (unK) cells from miscarriage decidua, regarding their cytokine profile and endometrial stromal cell (ESC) crosstalk? SUMMARY ANSWER uNK-conditioned media from miscarriage samples present high TNF-α levels which inhibit ESC decidualisation. WHAT IS KNOWN ALREADY AEA plasma levels are higher in women who have suffered a miscarriage. Moreover, AEA inhibits ESC proliferation and differentiation, although the levels and impact on the uNK cell cytokine profile at the feto-maternal interface remain elusive. STUDY DESIGN, SIZE, DURATION This laboratory-based study used human primary uNK cells which were isolated from first-trimester decidua (gestational age, 5-12 weeks) derived from 8 women with elective pregnancy termination and 18 women who suffered a miscarriage. PARTICIPANTS/MATERIALS, SETTING, METHODS The first-trimester placental tissues were assayed for AEA levels by UPLC-MS/MS and respective enzymatic profile by western blot. The uNK cells were isolated and maintained in culture. The expression of angiogenic markers in uNK cells was examined by quantitative PCR (qPCR). The uNK-conditioned medium was analysed for IFN-γ, TNF-α and IL-10 production by enzyme-linked immunosorbent assay, and the impact on ESC differentiation was assessed by measuring decidual markers Prl, Igfbp-1 and Fox01 mRNA expression using qPCR. MAIN RESULTS AND THE ROLE OF CHANCE AEA levels were higher in miscarriage decidua compared with decidua from elective terminations. The uNK cell-conditioned medium from the miscarriage samples exhibited high TNF-α levels and interfered with the decidualisation of ESCs. Exacerbated inflammation and elevated TNF-α levels at the feto-maternal interface may trigger AEA signalling pathways that, in turn, may impact decidualisation and the angiogenic ability of uNK cells. LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Primary uNK cell responses are based on a simple in vitro model. Thus, in complex microenvironments, such as the feto-maternal interface, the mechanisms may not be exactly the same. Also, the inflammatory events of miscarriage that, in this study, have happened prior to processing of the samples may cause different responses to that observed. In addition, the magnitude of the inflammatory response, required to trigger the AEA pathways that impact decidualisation and the uNK angiogenic ability in vivo, is still unclear. WIDER IMPLICATIONS OF THE FINDINGS The endocannabinoid AEA is a modulator of reproductive competence. AEA not only may contribute to neuroendocrine homeostasis but also can take part in uterine changes occurring during early pregnancy. STUDY FUNDING/COMPETING INTEREST(S) The work was supported by UID/MULTI/04378/2019 with funding from Fundação para a Ciência e a Tecnologia (FCT)/MCTES through national funds and PORTUGAL 2020 Partnership Agreement, NORTE-01-0145-FEDER-000024. S.C. Cunha acknowledges FCT for the IF/01616/2015 contract. There are no conflicts of interest.
Collapse
Affiliation(s)
- B M Fonseca
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - S C Cunha
- LAQV, REQUIMTE, Laboratório de Bromatologia e Hidrologia, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - D Gonçalves
- Departamento da Mulher e da Medicina Reprodutiva, Serviço de Obstetrícia, Centro Materno-Infantil do Norte, Centro Hospitalar do Porto, Porto, Portugal
| | - A Mendes
- Departamento da Mulher e da Medicina Reprodutiva, Serviço de Obstetrícia, Centro Materno-Infantil do Norte, Centro Hospitalar do Porto, Porto, Portugal
| | - J Braga
- Departamento da Mulher e da Medicina Reprodutiva, Serviço de Obstetrícia, Centro Materno-Infantil do Norte, Centro Hospitalar do Porto, Porto, Portugal
| | - G Correia-da-Silva
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - N A Teixeira
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| |
Collapse
|
38
|
Zhang Z, Zeng P, Gao W, Wu R, Deng T, Chen S, Tian X. Exploration of the Potential Mechanism of Calculus Bovis in Treatment of Primary Liver Cancer by Network Pharmacology. Comb Chem High Throughput Screen 2021; 24:129-138. [PMID: 32772910 DOI: 10.2174/1386207323666200808172051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/21/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022]
Abstract
AIM AND OBJECTIVE Calculus Bovis (CB) has been employed to treat diseases for a long time. It has been identified to play significant anti-inflammatory and anti-tumor roles. However, the mechanism of treating primary liver cancer (PLC) remains to be revealed. This study aims to clarify the molecules and mechanisms of CB in treating PLC. MATERIALS AND METHODS After oral bioavailability (OB) and drug-likeness (DL) screening, 15 small molecules were identified as the potential ingredients against PLC. Following this, related targets network constructions and pathways were applied to clarify the mechanism of CB in treating PLC. An in vitro experiment was carried out to identify the function of CB in treating PLC. RESULTS Eleven compounds of CB were identified that play an anti-PLC role, including oleanolic acid, ergosterol, ursolic acid, etc. The potential targets which were observed include IL6, MAPK-8, VEGFA, Caspase-3, etc. Further analysis showed that the mechanism of CB in the treatment of PLC involved apoptosis-related pathways and immune-related pathways. CONCLUSION In summary, the current study combines network pharmacology and in vitro experiments to reveal the mechanism of CB against PLC. We concluded that 11 ingredients of CB have an anti-PLC effect. Furthermore, CB plays a key role in treating PLC mainly by apoptosisrelated pathways and immune-related pathways. Our experiment verifies that CB promotes the apoptosis of SMMC-7721.
Collapse
Affiliation(s)
- Zhen Zhang
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Puhua Zeng
- Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha 410006, China
| | - Wenhui Gao
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ruoxia Wu
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Tianhao Deng
- Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha 410006, China
| | - Siqin Chen
- Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha 410006, China
| | - Xuefei Tian
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
39
|
Gallardo Paffetti M, Cárcamo J, Arias-Darraz L, Alvear C, Ojeda J. Effect of Type of Pregnancy on Transcriptional and Plasma Metabolic Response in Sheep and Its Further Effect on Progeny Lambs. Animals (Basel) 2020; 10:ani10122290. [PMID: 33287438 PMCID: PMC7761827 DOI: 10.3390/ani10122290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The present study was carried out in order to determine the effect of type of pregnancy on the mammary gland development, evaluated through the transcriptional expression of genes that are associated to angiogenesis and cell turnover/lactogenesis and the metabolic response of the animals. For this, six twin and seven single-bearing ewes were fed with naturalized pasture from day −45 pre-partum until day +70 post-partum, taking samples of mammary tissue and plasma at different times from the birth until weaning. The results showed the type of pregnancy could only explain a few differences in the transcriptional expression of in some genes that are involved in angiogenesis and cell turnover/lactogenesis in the mammary gland tissue, which had no impact on the metabolic status of ewes or the metabolic response in plasma, performance, and muscle transcriptional expression of the lambs. Abstract The following study was performed in order to determine the effect of type of pregnancy on the transcriptional expression of genes that are engaged in angiogenesis and cell turnover/lactogenesis in the ewe mammary gland, evaluating its impact on the plasma metabolic response. In addition, an assessment of its further influence on plasma metabolic response, performance, and muscle transcriptional expression of lipogenic enzymes in progeny lambs was made. Thirteen Ile de France sheep (six twin- and seven single-bearing ewes) were allocated to graze ad libitum naturalized pasture from d 45 pre-partum to day 70 post-partum, while keeping their lambs on the same diet until day 60 after weaning. The samples were collected at different times and analyzed by qRT-PCR and plasma metabolic indicators. The data were processed using SPSS package. The results showed that twin-bearing ewes overexpressed VEGFR1 at birth, and BCL2 at birth and day 35 post-partum; however, single-bearing ewes overexpressed CAIV and IGF1 at day 35 post-partum. Similar metabolite concentrations in blood plasma were found between groups of ewes. The plasma metabolic response in lambs was similar between groups and it did not influence their performance, where a similar transcriptional expression of lipogenic enzymes in muscle was observed. Therefore, the type of pregnancy can explain the slight differences in mRNA expression that were found in angiogenesis and cell turnover/lactogenesis in mammary gland, although these differences not only did not affect the plasma metabolic response in ewes, but they also had no influence on plasma metabolic response, performance, and muscle transcriptional expression of their lambs.
Collapse
Affiliation(s)
- María Gallardo Paffetti
- Escuela de Medicina Veterinaria, Facultad de Ciencias, Universidad Mayor, Santiago PO Box 8580745, Chile; (M.G.P.); (C.A.)
| | - Juan Cárcamo
- Centro FONDAP, Interdisciplinary Center for Aquaculture Research (INCAR), Facultad de Ciencias, Universidad Austral de Chile, Valdivia PO Box 567, Chile;
- Correspondence: ; Tel.: +56-632-293-413
| | - Luis Arias-Darraz
- Centro FONDAP, Interdisciplinary Center for Aquaculture Research (INCAR), Facultad de Ciencias, Universidad Austral de Chile, Valdivia PO Box 567, Chile;
| | - Carlos Alvear
- Escuela de Medicina Veterinaria, Facultad de Ciencias, Universidad Mayor, Santiago PO Box 8580745, Chile; (M.G.P.); (C.A.)
| | - Javier Ojeda
- Instituto de Ciencias Clínicas Veterinarias, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia PO Box 567, Chile;
| |
Collapse
|
40
|
Kikuchi R, Tsuboi N, Sada KE, Nakatochi M, Yokoe Y, Suzuki A, Maruyama S, Murohara T, Matsushita T, Amano K, Atsumi T, Takasaki Y, Ito S, Hasegawa H, Dobashi H, Ito T, Makino H, Matsuo S. Vascular endothelial growth factor (VEGF)-A and VEGF-A 165b are associated with time to remission of granulomatosis with polyangiitis in a nationwide Japanese prospective cohort study. Ann Clin Biochem 2020; 58:86-94. [PMID: 33081494 DOI: 10.1177/0004563220968371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Effective prognostic markers are needed for antineutrophil cytoplasmic antibody-associated vasculitis (AAV). This study evaluated the clinical associations of serum vascular endothelial growth factor-A (sVEGF-A) and sVEGF-A165b (an antiangiogenic isoform of VEGF-A) concentrations with time to remission of AAV in a nationwide Japanese prospective follow-up cohort. METHODS We collected samples from patients with AAV who were enrolled in the nationwide Japanese cohort study (RemIT-JAV-RPGN). We measured sVEGF-A and sVEGF-A165b concentrations using enzyme-linked immunosorbent assays in 57 serum samples collected 6 months before and after initiation of AAV treatment. Patients were classified based on AAV disease subtypes: microscopic polyangiitis, granulomatosis with polyangiitis and eosinophilic granulomatosis with polyangiitis (EGPA). RESULTS Results revealed significant reductions in sVEGF-A and sVEGF-A165b concentrations in patients with microscopic polyangiitis and EGPA, respectively. However, despite the comparable concentrations of sVEGF-A and sVEGF-A165b during the 6 months of treatment in granulomatosis with polyangiitis patients, correlation analysis revealed that the differences in log2-transformed concentrations of sVEGF-A and sVEGF-A165b were inversely correlated with time to remission in granulomatosis with polyangiitis patients. CONCLUSION These results suggest that sVEGF-A and -A165b can serve as potential markers of time to remission in patients with granulomatosis with polyangiitis.
Collapse
Affiliation(s)
- Ryosuke Kikuchi
- Department of Medical Technique, Nagoya University Hospital, Nagoya, Japan
| | - Naotake Tsuboi
- Department of Nephrology, School of Medicine, Fujita Health University, Nagoya, Japan
| | - Ken-Ei Sada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Clinical Epidemiology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Masahiro Nakatochi
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Yokoe
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsuo Suzuki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tadashi Matsushita
- Department of Clinical Laboratory, Nagoya University Hospital, Nagoya, Japan.,Department of Transfusion Medicine, Nagoya University Hospital, Nagoya, Japan
| | | | - Koichi Amano
- Department of Rheumatology and Clinical Immunology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Tatsuya Atsumi
- Rheumatology, Endocrinology and Nephrology, Faculty of Medicine, Hokkaido University, Hokkaido, Japan
| | - Yoshinari Takasaki
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Satoshi Ito
- Department of Rheumatology, Niigata Rheumatic Center, Niigata, Japan
| | - Hitoshi Hasegawa
- Department of Hematology, Clinical Immunology, and Infectious Diseases, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroaki Dobashi
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takafumi Ito
- Division of Nephrology, Shimane University, Faculty of Medicine, Shimane, Japan
| | | | | |
Collapse
|
41
|
Wang X, Bove AM, Simone G, Ma B. Molecular Bases of VEGFR-2-Mediated Physiological Function and Pathological Role. Front Cell Dev Biol 2020; 8:599281. [PMID: 33304904 PMCID: PMC7701214 DOI: 10.3389/fcell.2020.599281] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022] Open
Abstract
The vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) play crucial roles in vasculogenesis and angiogenesis. Angiogenesis is an important mechanism in many physiological and pathological processes, and is involved in endothelial cell proliferation, migration, and survival, then leads to further tubulogenesis, and finally promotes formation of vessels. This series of signaling cascade pathways are precisely mediated by VEGF/VEGFR-2 system. The VEGF binding to the IgD2 and IgD3 of VEGFR-2 induces the dimerization of the receptor, subsequently the activation and trans-autophosphorylation of the tyrosine kinase, and then the initiation of the intracellular signaling cascades. Finally the VEGF-activated VEGFR-2 stimulates and mediates variety of signaling transduction, biological responses, and pathological processes in angiogenesis. Several crucial phosphorylated sites Tyr801, Try951, Try1175, and Try1214 in the VEGFR-2 intracellular domains mediate several key signaling processes including PLCγ-PKC, TSAd-Src-PI3K-Akt, SHB-FAK-paxillin, SHB-PI3K-Akt, and NCK-p38-MAPKAPK2/3 pathways. Based on the molecular structure and signaling pathways of VEGFR-2, the strategy of the VEGFR-2-targeted therapy should be considered to employ in the treatment of the VEGF/VEGFR-2-associated diseases by blocking the VEGF/VEGFR-2 signaling pathway, inhibiting VEGF and VEGFR-2 gene expression, blocking the binding of VEGF and VEGFR-2, and preventing the proliferation, migration, and survival of vascular endothelial cells expressing VEGFR-2.
Collapse
Affiliation(s)
- Xinrong Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | | | | | - Binyun Ma
- Department of Medicine/Hematology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
42
|
Choi MA, Saeidi S, Han HJ, Kim SJ, Kwon N, Kim DH, Min SH, Choi BY, Surh YJ. The peptidyl prolyl isomerase, PIN1 induces angiogenesis through direct interaction with HIF-2α. Biochem Biophys Res Commun 2020; 533:995-1003. [PMID: 33012513 DOI: 10.1016/j.bbrc.2020.08.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/27/2022]
Abstract
PIN1, the peptidyl-prolyl isomerase (PPIase), is an enzyme that changes the conformation of phosphoproteins. The conformational change induced by PIN1 alters the function and stability of the target proteins. PIN1 is overexpressed in many different types of malignancies, including breast, lung, cervical, brain and colorectal tumors. PIN1 overexpression has been associated with activation of multiple oncogenic signaling pathways during tumor development. Hypoxia-inducible factor 2α (HIF-2α), a transcription factor activated in hypoxia, plays a role in erythropoiesis, glycolysis, tissue invasion, metastasis and angiogenesis. In this study, we found the direct interaction between HIF-2α and PIN1 in colorectal cancer HCT116 cells. Notably, serine 16 and lysine 63 residues of PIN1 were critical for its interaction with HIF-2α. When PIN1 protein was silenced by transient transfection of PIN1 short interfering RNA, the expression of HIF-2α was attenuated under a hypoxic condition. Moreover, genetic and pharmacologic inhibition of PIN1 abrogated the expression of vascular endothelial growth factor and angiogenesis. The cycloheximide chase experiment revealed the stabilization of HIF-2α by PIN1. Both WW and PPIase domains of PIN1 appear to be critical for its interaction with HIF-2α.
Collapse
Affiliation(s)
- Min-A Choi
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Soma Saeidi
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Hyeong-Jun Han
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Su-Jung Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Nayoung Kwon
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do, 16227, South Korea
| | - Sang-Hyun Min
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, 701-310, South Korea
| | - Bu Young Choi
- Department of Pharmaceutical Science and Engineering, School of Convergence Bioscience and Technology, Seowon University, Chungbuk, 28674, South Korea
| | - Young-Joon Surh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea; Cancer Research Institute, Seoul National University, Seoul, 03080, South Korea.
| |
Collapse
|
43
|
Rodriguez AE, Gisbert S, Palazón A, Alio JL. Quantification of Growth Factors and Fibronectin in Diverse Preparations of Platelet-Rich Plasma for the Treatment of Ocular Surface Disorders (E-PRP). Transl Vis Sci Technol 2020; 9:22. [PMID: 32821519 PMCID: PMC7409215 DOI: 10.1167/tvst.9.6.22] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 03/26/2020] [Indexed: 01/11/2023] Open
Abstract
Purpose The purpose of this study was to quantify the presence of growth factors (GFs) and fibronectin in autologous platelet-rich plasma for topical ocular use (E-PRP) comparing their concentration when different preparation and preservation procedures were applied. Methods E-PRP was prepared with blood from healthy volunteers. The count of platelets, leukocytes, and red blood cells in the whole blood and E-PRP were performed. The concentration of the GFs platelet-derived growth factor BB (PDGF-BB), transforming growth factor β1 (TGF-β1), epidermal growth factor (EGF), vascular endothelial growth factor A (VEGF-A), and fibronectin was determined in each of the four procedures applied including fresh, frozen at −20°C for 3 months, fresh-spin, and frozen-spin at −20°C E-PRP samples. Posterior statistical analysis was performed to establish significant differences between groups and between GFs in relation to the amounts of platelets. Results Platelets in the E-PRP doubled in the number of basal values of whole blood (P ≤ 0.01). The blood cells in the E-PRP decreased drastically in red cells (99%) and also in leukocytes (82%). The concentration of PDGF-BB and EGF was significantly higher (P < 0.01) when the E-PRP samples were frozen at −20°C. However, no significant differences were observed for TGF-β1, VEGF-A, and fibronectin (P > 0.05). The concentration of GFs in the E-PRP did not necessarily correlate with the number of platelets. Conclusions Freezing the E-PRP for 3 months at −20°C increased the concentration of important proteins, such as PDGF-BB and EGF, and maintained the levels of others. These findings are essential because treatments, such as E-PRP, used by patients with ocular surface dysfunctions tend to prolong it in time. In addition, subsequent centrifugation of the E-PRP decreased the values of TFG-β1, but not the other GFs, which would allow adjusting the concentration of TFG-β1, as necessary. This procedure guarantees their correct conservation and viability. Translational Relevance This work demonstrates how clinical application can be improved by starting from basic research. The quantification of GFs and fibronectin in platelet-rich plasma (PRP) helps to clarify which is the best mode of preparation and preservation of PRP for clinical applications. This allows to optimize the product that is delivered to the patients as a treatment for the dysfunctions of the ocular surface, guaranteeing that the conservation does not affect at all the quality of the PRP that it is going to be used.
Collapse
Affiliation(s)
- Alejandra E Rodriguez
- Research and Development Laboratory, Vissum Miranza, Alicante, Spain.,Department of Cornea and Refractive Surgery, Vissum Miranza, Alicante, Spain.,Department of Ophthalmology, Miguel Hernandez University, Alicante, Spain
| | - Sandra Gisbert
- Research and Development Laboratory, Vissum Miranza, Alicante, Spain
| | - Antonio Palazón
- Department of Clinical Medicine, Miguel Hernandez University, Alicante, Spain
| | - Jorge L Alio
- Department of Cornea and Refractive Surgery, Vissum Miranza, Alicante, Spain.,Department of Ophthalmology, Miguel Hernandez University, Alicante, Spain
| |
Collapse
|
44
|
Jena MK, Sharma NR, Petitt M, Maulik D, Nayak NR. Pathogenesis of Preeclampsia and Therapeutic Approaches Targeting the Placenta. Biomolecules 2020; 10:biom10060953. [PMID: 32599856 PMCID: PMC7357118 DOI: 10.3390/biom10060953] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/20/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
Preeclampsia (PE) is a serious pregnancy complication, affecting about 5–7% of pregnancies worldwide and is characterized by hypertension and damage to multiple maternal organs, primarily the liver and kidneys. PE usually begins after 20 weeks’ gestation and, if left untreated, can lead to serious complications and lifelong disabilities—even death—in both the mother and the infant. As delivery is the only cure for the disease, treatment is primarily focused on the management of blood pressure and other clinical symptoms. The pathogenesis of PE is still not clear. Abnormal spiral artery remodeling, placental ischemia and a resulting increase in the circulating levels of vascular endothelial growth factor receptor-1 (VEGFR-1), also called soluble fms-like tyrosine kinase-1 (sFlt-1), are believed to be among the primary pathologies associated with PE. sFlt-1 is produced mainly in the placenta during pregnancy and acts as a decoy receptor, binding to free VEGF (VEGF-A) and placental growth factor (PlGF), resulting in the decreased bioavailability of each to target cells. Despite the pathogenic effects of increased sFlt-1 on the maternal vasculature, recent studies from our laboratory and others have strongly indicated that the increase in sFlt-1 in PE may fulfill critical protective functions in preeclamptic pregnancies. Thus, further studies on the roles of sFlt-1 in normal and preeclamptic pregnancies are warranted for the development of therapeutic strategies targeting VEGF signaling for the treatment of PE. Another impediment to the treatment of PE is the lack of suitable methods for delivery of cargo to placental cells, as PE is believed to be of placental origin and most available therapies for PE adversely impact both the mother and the fetus. The present review discusses the pathogenesis of PE, the complex role of sFlt-1 in maternal disease and fetal protection, and the recently developed placenta-targeted drug delivery system for the potential treatment of PE with candidate therapeutic agents.
Collapse
Affiliation(s)
- Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Phagwara, Punjab 144411, India;
- Correspondence:
| | - Neeta Raj Sharma
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Phagwara, Punjab 144411, India;
| | - Matthew Petitt
- Redwood Biomedical Editing, Redwood City, CA 94061, USA;
| | - Devika Maulik
- Department of Obstetrics and Gynecology, UMKC School of Medicine, Kansas City, MO 64108, USA; (D.M.); (N.R.N.)
| | - Nihar Ranjan Nayak
- Department of Obstetrics and Gynecology, UMKC School of Medicine, Kansas City, MO 64108, USA; (D.M.); (N.R.N.)
| |
Collapse
|
45
|
Dong T, Duan C, Wang S, Gao X, Yang Q, Yang W, Deng Y. Multifunctional Surface with Enhanced Angiogenesis for Improving Long-Term Osteogenic Fixation of Poly(ether ether ketone) Implants. ACS APPLIED MATERIALS & INTERFACES 2020; 12:14971-14982. [PMID: 32159330 DOI: 10.1021/acsami.0c02304] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Poly(ether ether ketone) (PEEK) is a biocompatible polymer, but the lack of angiogenesis makes the long-term osteogenic fixation of PEEK implants challenging, which has hampered their wider application in orthopedics. Herein, we develop a multifunctional micro-/nanostructured surface presenting hydroxyapatite (HA) nanoflowers and nickel hydroxide (Ni(OH)2) nanoparticles on PEEK implants (sPEEK-Ni-HA) to tackle the problem. The results show that the reasonable release of Ni2+ from sPEEK-Ni-HA significantly facilitates the migration, tube formation, and angiogenic gene expression of human umbilical vein endothelial cells (HUVECs). In addition to angiogenesis, the sPEEK-Ni-HA displays enhanced cytocompatibility and osteogenicity in terms of cell proliferation, spreading, alkaline phosphatase activity, matrix mineralization, and osteogenesis-related gene secretion, exceeding pure and other multifunctional sPEEK samples. Importantly, in vivo evaluations employing a rabbit femoral condyle implantation model confirm that such dual decoration of Ni elements and HA nanoflowers boosts bone remodeling/osseointegration, which dramatically promotes the in vivo osteogenic fixation of implants. Therefore, this work not only sheds light on the significance of angiogenesis on the osteogenic fixation of an implant but also presents a facile strategy to empower bioinert PEEK with a well-orchestrated feature of angiogenesis and osteogenesis.
Collapse
Affiliation(s)
| | - Chunyan Duan
- School of Basic Medical Science, Southwest Medical University, Luzhou 64600, China
| | - Song Wang
- Department of Spine Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou 64600, China
| | | | | | | | - Yi Deng
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
46
|
Resistance of melanoma cells to anticancer treatment: a role of vascular endothelial growth factor. Postepy Dermatol Alergol 2020; 37:11-18. [PMID: 32467677 PMCID: PMC7247075 DOI: 10.5114/ada.2020.93378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/21/2019] [Indexed: 12/18/2022] Open
Abstract
Melanoma is one of the most aggressive and resistant to treatment neoplasms. There are still many challenges despite many promising advances in anticancer treatment. Currently, the main problem for all types of treatment is associated with heterogeneity. Due to heterogeneity of cancer cells, "precise" targeting of a medicine against a single phenotype limits the efficacy of treatment and affects resistance to applied therapy. Therefore it is important to understand aetiology and reasons for heterogeneity in order to develop effective and long-lasting treatment. This review summarises roles of vascular endothelial growth factor (VEGF) that may stimulate growth of a melanoma tumour irrespective of its proangiogenic effects, contributing to cancer heterogeneity. VEGF triggers processes associated with extracellular matrix remodelling, cell migration, invasion, angiogenesis, inhibition of immune responses and favours phenotypic plasticity and epithelial-mesenchymal transition. Consequently, it participates in mechanisms of interactions between melanoma cancer cells and microenvironment and it can modify sensitivity to therapeutic factors.
Collapse
|
47
|
Wei P, Zhang Z, Lin M, Zhou B, Wang Z. Bevacizumab has bidirectional regulatory effects on the secretion of basic fibroblast growth factor in glioma cells. Cytokine 2020; 129:155022. [PMID: 32044668 DOI: 10.1016/j.cyto.2020.155022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 11/16/2022]
Abstract
Previous studies suggest that upregulated basic fibroblast growth factor (bFGF) plays a key role in the resistance to anti-vascular endothelial growth factor (VEGF) therapy in glioma. This study reported that anti-VEGF treatment regulated bFGF secretion in a double-edged manner. That is, moderate VEGF neutralization reduced bFGF production, whereas VEGF overblocking enhanced bFGF secretion in glioma cells. Our data provide a new perspective on the treatment of glioma with anti-VEGF, and the underlying mechanism is worthy of further study.
Collapse
Affiliation(s)
- Pei Wei
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Zhainan Zhang
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Mao Lin
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Bin Zhou
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Zhiyong Wang
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
48
|
Bai C, Sun Y, Pan X, Yang J, Li X, Wu A, Qin D, Cao S, Zou W, Wu J. Antitumor Effects of Trimethylellagic Acid Isolated From Sanguisorba officinalis L. on Colorectal Cancer via Angiogenesis Inhibition and Apoptosis Induction. Front Pharmacol 2020; 10:1646. [PMID: 32047442 PMCID: PMC6997556 DOI: 10.3389/fphar.2019.01646] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
Previous studies have demonstrated that tannin could inhibit the proliferation and angiogenesis of cancer cells. However, the mechanism(s) associated with its antitumor effect remains unclear. Here, we investigated the effects of 3,3',4'-trimethylellagic acid (TMEA), a tannin compound isolated from Sanguisorba officinalis L., on the proliferation, angiogenesis, and apoptosis in cancer cells, as well as the underlying mechanism(s) related to its antitumor activity. TMEA was isolated from Sanguisorba officinalis L. by silica gel column chromatography. Molecular docking was carried out to assess active pocket binding between TMEA and vascular endothelial growth factor receptor 2 (VEGFR2). The antiangiogenic effect of TMEA on the migration and tube formation was detected in HUVECs by wound healing and tube formation assays, respectively. The antitumor effects of TMEA on the cell proliferation were determined in HepG2, A549, and SW620 cells by MTS assay in vitro and on the tumor growth of SW620 xenografts bearing in nude mice in vivo. The mRNA expression of Bcl-2, Bax, caspase-3, VEGF, PI3K, and mTOR were measured by qRT-PCR and protein expression of Bcl-2, Bax, caspase-3, VEGF, PI3K, and mTOR by Western blotting, and the protein expression of Bcl-2, Bax, caspase-3 and CD31 were detected by immunohistochemical analysis in vivo, respectively. The results showed that TMEA combined with VEGFR2 in the functional pockets of Asn223A, Gly922A, and Leu840A and inhibited the proliferation, migration, tube formation, and expression of VEGF and its downstream signaling mediators in HUVECs. TMEA also significantly inhibited the proliferation of HepG2, A549, and SW620 cancer cells in vitro, and suppressed the growth of SW620 tumors in vivo. Moreover, TMEA upregulated the expression of proapoptotic factors Bax and caspase-3 and downregulated the expression of antiapoptotic factors CD31 and Bcl-2 in cancer cells and/or tumor tissues. The data indicate that TMEA executes its anticancer activity by inducing apoptosis and inhibiting angiogenesis in cancer cells in vitro and tumor growth in vivo. The underlying anticancer mechanism is associated with the apoptotic and VEGF/PI3K/AKT/mTOR pathways.
Collapse
Affiliation(s)
- Chongfei Bai
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueshan Sun
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xianchao Pan
- Department of Medicine, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Yang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Institute of Cardiovascular Research, Key Laboratory of Medical Electrophysiology, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Xiaoxuan Li
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Institute of Cardiovascular Research, Key Laboratory of Medical Electrophysiology, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Dalian Qin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Institute of Cardiovascular Research, Key Laboratory of Medical Electrophysiology, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wenjun Zou
- Department of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianming Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Institute of Cardiovascular Research, Key Laboratory of Medical Electrophysiology, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
49
|
Taylor OG, Brzozowski JS, Skelding KA. Glioblastoma Multiforme: An Overview of Emerging Therapeutic Targets. Front Oncol 2019; 9:963. [PMID: 31616641 PMCID: PMC6775189 DOI: 10.3389/fonc.2019.00963] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/11/2019] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive malignant primary brain tumour in humans and has a very poor prognosis. The existing treatments have had limited success in increasing overall survival. Thus, identifying and understanding the key molecule(s) responsible for the malignant phenotype of GBM will yield new potential therapeutic targets. The treatment of brain tumours faces unique challenges, including the presence of the blood brain barrier (BBB), which limits the concentration of drugs that can reach the site of the tumour. Nevertheless, several promising treatments have been shown to cross the BBB and have shown promising pre-clinical results. This review will outline the status of several of these promising targeted therapies.
Collapse
Affiliation(s)
- Olivia G Taylor
- Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Cancer Research Alliance and Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Joshua S Brzozowski
- Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Cancer Research Alliance and Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Kathryn A Skelding
- Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Cancer Research Alliance and Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
50
|
Wallace B, Peisl A, Seedorf G, Nowlin T, Kim C, Bosco J, Kenniston J, Keefe D, Abman SH. Anti-sFlt-1 Therapy Preserves Lung Alveolar and Vascular Growth in Antenatal Models of Bronchopulmonary Dysplasia. Am J Respir Crit Care Med 2019; 197:776-787. [PMID: 29268623 DOI: 10.1164/rccm.201707-1371oc] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
RATIONALE Pregnancies complicated by antenatal stress, including preeclampsia (PE) and chorioamnionitis (CA), increase the risk for bronchopulmonary dysplasia (BPD) in preterm infants, but biologic mechanisms linking prenatal factors with BPD are uncertain. Levels of sFlt-1 (soluble fms-like tyrosine kinase 1), an endogenous antagonist to VEGF (vascular endothelial growth factor), are increased in amniotic fluid and maternal blood in PE and associated with CA. OBJECTIVES Because impaired VEGF signaling has been implicated in the pathogenesis of BPD, we hypothesized that fetal exposure to sFlt-1 decreases lung growth and causes abnormal lung structure and pulmonary hypertension during infancy. METHODS To test this hypothesis, we studied the effects of anti-sFlt-1 monoclonal antibody (mAb) treatment on lung growth in two established antenatal models of BPD that mimic PE and CA induced by intraamniotic (i.a.) injections of sFlt-1 or endotoxin, respectively. In experimental PE, mAb was administered by three different approaches, including antenatal treatment by either i.a. instillation or maternal uterine artery infusion, or by postnatal intraperitoneal injections. RESULTS With each strategy, mAb therapy improved infant lung structure as assessed by radial alveolar count, vessel density, right ventricular hypertrophy, and lung function. As found in the PE model, the adverse lung effects of i.a. endotoxin were also reduced by antenatal or postnatal mAb therapy. CONCLUSIONS We conclude that treatment with anti-sFlt-1 mAb preserves lung structure and function and prevents right ventricular hypertrophy in two rat models of BPD of antenatal stress and speculate that early mAb therapy may provide a novel strategy for the prevention of BPD.
Collapse
Affiliation(s)
| | | | - Gregory Seedorf
- 1 Pediatric Heart Lung Center.,3 Department of Pediatrics, University of Colorado Denver Anschutz Medical Center and Children's Hospital Colorado, Aurora, Colorado; and
| | - Taylor Nowlin
- 1 Pediatric Heart Lung Center.,3 Department of Pediatrics, University of Colorado Denver Anschutz Medical Center and Children's Hospital Colorado, Aurora, Colorado; and
| | - Christina Kim
- 1 Pediatric Heart Lung Center.,2 Department of Surgery, and
| | | | | | - Dennis Keefe
- 4 Shire Pharmaceuticals, Lexington, Massachusetts
| | - Steven H Abman
- 1 Pediatric Heart Lung Center.,3 Department of Pediatrics, University of Colorado Denver Anschutz Medical Center and Children's Hospital Colorado, Aurora, Colorado; and
| |
Collapse
|