1
|
You Y, Tian Y, Guo R, Shi J, Kwak KJ, Tong Y, Estania AP, Hsu WH, Liu Y, Hu S, Cao J, Yang L, Bai R, Huang P, Lee LJ, Jiang W, Kim BYS, Ma S, Liu X, Shen Z, Lan F, Phuong Nguyen PK, Lee AS. Extracellular vesicle-mediated VEGF-A mRNA delivery rescues ischaemic injury with low immunogenicity. Eur Heart J 2025:ehae883. [PMID: 39831819 DOI: 10.1093/eurheartj/ehae883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/09/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND AIMS Lackluster results from recently completed gene therapy clinical trials of VEGF-A delivered by viral vectors have heightened the need to develop alternative delivery strategies. This study aims to demonstrate the pre-clinical efficacy and safety of extracellular vesicles (EVs) loaded with VEGF-A mRNA for the treatment of ischaemic vascular disease. METHODS After encapsulation of full-length VEGF-A mRNA into fibroblast-derived EVs via cellular nanoporation (CNP), collected VEGF-A EVs were delivered into mouse models of ischaemic injury. Target tissue delivery was verified by in situ analysis of protein and gene expression. Functional rescue was confirmed by in vivo imaging and histology. The safety of single and serial delivery was demonstrated using immune-based assays. RESULTS VEGF-A EVs were generated with high mRNA content using a CNP methodology. VEGF-A EV administration demonstrated expression of exogenous VEGF-A mRNA by in situ RNA hybridization and elevated protein expression by western blot, microscopy, and enzyme-linked immunosorbent assay. Mice treated with human VEGF-A EVs after femoral or coronary artery ligation exhibited heightened neovascularization in ischaemic tissues with increased arterial perfusion and improvement in left ventricular function, respectively. Serial delivery of VEGF-EVs in injured skin showed improved wound healing with repeat administration. Importantly, as compared with adeno-associated viral and lipid nanoparticle VEGF-A gene therapy modalities, murine VEGF-A EV delivery did not trigger innate or adaptive immune responses at the injection site or systemically. CONCLUSIONS This study demonstrated that VEGF-A EV therapy offers efficient, dose-dependent VEGF-A protein formation with low immunogenicity, resulting in new vessel formation in murine models of ischaemic vascular disease.
Collapse
Affiliation(s)
- Yi You
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Yu Tian
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Rui Guo
- Department of Cardiac Surgery, Peking University Third Hospital, 49 Huayuan N Rd, Haidian District, Beijing 100191, China
| | - Junfeng Shi
- Department of Chemical and Biomolecular Engineering, 151 W Woodruff Ave, Columbus, The Ohio State University, Columbus, OH 43210, USA
| | - Kwang Joo Kwak
- Department of Chemical and Biomolecular Engineering, 151 W Woodruff Ave, Columbus, The Ohio State University, Columbus, OH 43210, USA
| | - Yuhao Tong
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Andreanne Poppy Estania
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Wei-Hsiang Hsu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Yutong Liu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Shijun Hu
- Department of Cardiovascular Surgery for the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, China
| | - Jianhong Cao
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Liqun Yang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Rui Bai
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
| | - Pufeng Huang
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
| | - Ly James Lee
- Spot Biosystems Ltd, 432 High Street, Apartment 201, Palo Alto, CA 94301, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX 77030, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Shuhong Ma
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
| | - Xujie Liu
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery for the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, China
| | - Feng Lan
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Research Institute Building, Room 323, 2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Patricia Kim Phuong Nguyen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, 870 Quarry Road, Rm 183, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, 265 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew S Lee
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
- Greater Bay Area International Clinical Trials Center, Shenzhen Medical Academy of Research and Translation, Shenzhen 518055, China
| |
Collapse
|
2
|
Zhao X, Ma C, Li L, Yang Y, Zhang S, Wang X. Human Adipose Tissue-Derived Stromal Cells Ameliorate Adriamycin-Induced Nephropathy by Promoting Angiogenesis. Organogenesis 2024; 20:2356339. [PMID: 38796830 PMCID: PMC11135856 DOI: 10.1080/15476278.2024.2356339] [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: 12/14/2023] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
This study is to investigate the therapeutical effect and mechanisms of human-derived adipose mesenchymal stem cells (ADSC) in relieving adriamycin (ADR)-induced nephropathy (AN). SD rats were separated into normal group, ADR group, ADR+Losartan group (20 mg/kg), and ADR + ADSC group. AN rats were induced by intravenous injection with adriamycin (8 mg/kg), and 4 d later, ADSC (2 × 105 cells/mouse) were administrated twice with 2 weeks interval time (i.v.). The rats were euthanized after the 6 weeks' treatment. Biochemical indicators reflecting renal injury, such as blood urea nitrogen (BUN), neutrophil gelatinase alpha (NGAL), serum creatinine (Scr), inflammation, oxidative stress, and pro-fibrosis molecules, were evaluated. Results demonstrated that we obtained high qualified ADSCs for treatment determined by flow cytometry, and ADSCs treatment significantly ameliorated renal injuries in DN rats by decreasing BUN, Scr and NGAL in peripheral blood, as well as renal histopathological injuries, especially protecting the integrity of podocytes by immunofluorescence. Furthermore, ADSCs treatment also remarkably reduced the renal inflammation, oxidative stress, and fibrosis in DN rats. Preliminary mechanism study suggested that the ADSCs treatment significantly increased renal neovascularization via enhancing proangiogenic VEGF production. Pharmacodynamics study using in vivo imaging confirmed that ADSCs via intravenous injection could accumulate into the kidneys and be alive at least 2 weeks. In a conclusion, ADSC can significantly alleviate ADR-induced nephropathy, and mainly through reducing oxidative stress, inflammation and fibrosis, as well as enhancing VEGF production.
Collapse
Affiliation(s)
- Xiaodi Zhao
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chengyan Ma
- The Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lijie Li
- Beijing AeglesStem Technology Co. LTD, Beijing, China
| | - Yuemei Yang
- Beijing AeglesStem Technology Co. LTD, Beijing, China
| | - Sen Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaoli Wang
- Department of Hematology, Lishui People’s Hospital, Lishui, China
| |
Collapse
|
3
|
Liang B, Zhou Y, Qin Y, Li X, Zhou S, Yuan K, Zhao R, Lv X, Qin D. Research Progress on Using Nanoparticles to Enhance the Efficacy of Drug Therapy for Chronic Mountain Sickness. Pharmaceutics 2024; 16:1375. [PMID: 39598498 PMCID: PMC11597246 DOI: 10.3390/pharmaceutics16111375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Chronic mountain sickness (CMS) poses a significant health risk to individuals who rapidly ascend to high altitudes, potentially endangering their lives. Nanoparticles (NPs) offer an effective means of transporting and delivering drugs, protecting nucleic acids from nuclease degradation, and mediating the expression of target genes in specific cells. These NPs are almost non-toxic and easy to prepare and store, possess a large surface area, exhibit good biocompatibility and degradability, and maintain good stability. They can be utilized in the treatment of CMS to enhance the therapeutic efficacy of drugs. This paper provides an overview of the impact of NPs on CMS, discussing their roles as nanocarriers and their potential in CMS treatment. It aims to present novel therapeutic strategies for the clinical management of CMS and summarizes the relevant pathways through which NPs contribute to plateau disease treatment, providing a theoretical foundation for future clinical research.
Collapse
Affiliation(s)
- Boshen Liang
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (B.L.); (Y.Z.); (Y.Q.); (X.L.); (S.Z.)
| | - Yang Zhou
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (B.L.); (Y.Z.); (Y.Q.); (X.L.); (S.Z.)
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming 650500, China;
| | - Yuliang Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (B.L.); (Y.Z.); (Y.Q.); (X.L.); (S.Z.)
| | - Xinyao Li
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (B.L.); (Y.Z.); (Y.Q.); (X.L.); (S.Z.)
| | - Sitong Zhou
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (B.L.); (Y.Z.); (Y.Q.); (X.L.); (S.Z.)
| | - Kai Yuan
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming 650500, China;
| | - Rong Zhao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming 650500, China;
| | - Xiaoman Lv
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (B.L.); (Y.Z.); (Y.Q.); (X.L.); (S.Z.)
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (B.L.); (Y.Z.); (Y.Q.); (X.L.); (S.Z.)
| |
Collapse
|
4
|
Wu M, Pokreisz P, Claus P, Casazza A, Gillijns H, Caluwé E, De Petrini M, Belmans A, Reyns G, Collen D, Janssens SP. Recombinant human placental growth factor-2 in post-infarction left ventricular dysfunction: a randomized, placebo-controlled, preclinical study. Basic Res Cardiol 2024; 119:795-806. [PMID: 39090343 DOI: 10.1007/s00395-024-01069-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 08/04/2024]
Abstract
Placental growth factor (PlGF)-2 induces angio- and arteriogenesis in rodents but its therapeutic potential in a clinically representative post-infarction left ventricular (LV) dysfunction model remains unclear. We, therefore, investigated the safety and efficacy of recombinant human (rh)PlGF-2 in the infarcted porcine heart in a randomized, placebo-controlled blinded study. We induced myocardial infarction (MI) in pigs using 75 min mid-LAD balloon occlusion followed by reperfusion. After 4 w, we randomized pigs with marked LV dysfunction (LVEF < 40%) to receive continuous intravenous infusion of 5, 15, 45 µg/kg/day rhPlGF-2 or PBS (CON) for 2 w using osmotic pumps. We evaluated the treatment effect at 8 w using comprehensive MRI and immunohistochemistry and measured myocardial PlGF-2 receptor transcript levels. At 4 w after MI, infarct size was 16-18 ± 4% of LV mass, resulting in significantly impaired systolic function (LVEF 34 ± 4%). In the pilot study (3 pigs/dose), PIGF administration showed sustained dose-dependent increases in plasma concentrations for 14 days without systemic toxicity and was associated with favorable post-infarct remodeling. In the second phase (n = 42), we detected no significant differences at 8 w between CON and PlGF-treated pigs in infarct size, capillary or arteriolar density, global LV function and regional myocardial blood flow at rest or during stress. Molecular analysis showed significant downregulation of the main PlGF-2 receptor, pVEGFR-1, in dysfunctional myocardium. Chronic rhPIGF-2 infusion was safe but failed to induce therapeutic neovascularization and improve global cardiac function after myocardial infarction in pigs. Our data emphasize the critical need for properly designed trials in representative large animal models before translating presumed promising therapies to patients.
Collapse
Affiliation(s)
- Ming Wu
- Department of Cardiovascular Sciences, KU Leuven, Campus Gasthuisberg, O&N1, 49 Herestraat, 3000, Leuven, Belgium
| | - Peter Pokreisz
- Department of Cardiovascular Sciences, KU Leuven, Campus Gasthuisberg, O&N1, 49 Herestraat, 3000, Leuven, Belgium
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- CoBioRes NV, Leuven, Belgium
| | - Piet Claus
- Department of Cardiovascular Sciences, KU Leuven, Campus Gasthuisberg, O&N1, 49 Herestraat, 3000, Leuven, Belgium
| | | | - Hilde Gillijns
- Department of Cardiovascular Sciences, KU Leuven, Campus Gasthuisberg, O&N1, 49 Herestraat, 3000, Leuven, Belgium
| | - Ellen Caluwé
- Department of Cardiovascular Sciences, KU Leuven, Campus Gasthuisberg, O&N1, 49 Herestraat, 3000, Leuven, Belgium
| | | | - Ann Belmans
- Leuven Biostatistics and Statistical Bioinformatics Center, KU Leuven, Leuven, Belgium
| | | | - Desire Collen
- Department of Cardiovascular Sciences, KU Leuven, Campus Gasthuisberg, O&N1, 49 Herestraat, 3000, Leuven, Belgium
- CoBioRes NV, Leuven, Belgium
| | - Stefan P Janssens
- Department of Cardiovascular Sciences, KU Leuven, Campus Gasthuisberg, O&N1, 49 Herestraat, 3000, Leuven, Belgium.
- Department of Cardiology, University Hospital Leuven, Leuven, Belgium.
| |
Collapse
|
5
|
Koteneva P, Kosheleva N, Fayzullin A, Khristidis Y, Rasulov T, Kulova A, Rozhkov S, Vedyaeva A, Brailovskaya T, Timashev P. Gene Therapeutic Drug pCMV-VEGF165 Plasmid ('Neovasculgen') Promotes Gingiva Soft Tissue Augmentation in Rabbits. Int J Mol Sci 2024; 25:10013. [PMID: 39337502 PMCID: PMC11432250 DOI: 10.3390/ijms251810013] [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: 08/09/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Currently, an increasing number of patients are undergoing extensive surgeries to restore the mucosa of the gums in the area of recessions. The use of a connective tissue graft from the palate is the gold standard of such surgical treatment, but complications, especially in cases of extensive defects, have led to the development of approaches using xenogeneic collagen matrices and methods to stimulate their regenerative and vasculogenic potential. This study investigated the potential of a xenogeneic scaffold derived from porcine skin Mucoderm and injections of the pCMV-VEGF165 plasmid ('Neovasculgen') to enhance soft gingival tissue volume and vascularization in an experimental model in rabbits. In vitro studies demonstrated the biocompatibility of the matrix and plasmid with gingival mesenchymal stem cells, showing no toxic effects and supporting cell viability and metabolic activity. In the in vivo experiment, the combination of Mucoderm and the pCMV-VEGF165 plasmid (0.12 mg) synergistically promoted tissue proliferation and vascularization. The thickness of soft tissues at the implantation site significantly increased with the combined application (3257.8 ± 1093.5 µm). Meanwhile, in the control group, the thickness of the submucosa was 341.8 ± 65.6 µm, and after the implantation of only Mucoderm, the thickness of the submucosa was 2041.6 ± 496.8 µm. Furthermore, when using a combination of Mucoderm and the pCMV-VEGF165 plasmid, the density and diameter of blood vessels were notably augmented, with a mean value of 226.7 ± 45.9 per 1 mm2 of tissue, while in the control group, it was only 68.3 ± 17.2 per 1 mm2 of tissue. With the application of only Mucoderm, it was 131.7 ± 37.1 per 1 mm2 of tissue, and with only the pCMV-VEGF165 plasmid, it was 145 ± 37.82 per 1 mm2 of the sample. Thus, the use of the pCMV-VEGF165 plasmid ('Neovasculgen') in combination with the xenogeneic collagen matrix Mucoderm potentiated the pro-proliferative effect of the membrane and the pro-vascularization effect of the plasmid. These results indicate the promising potential of this innovative approach for clinical applications in regenerative medicine and dentistry.
Collapse
Affiliation(s)
- Polina Koteneva
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Nastasia Kosheleva
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Alexey Fayzullin
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Yana Khristidis
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Timur Rasulov
- Central Research Institute of Dentistry and Maxillofacial Surgery, 119991 Moscow, Russia
| | - Aida Kulova
- Central Research Institute of Dentistry and Maxillofacial Surgery, 119991 Moscow, Russia
| | | | - Anna Vedyaeva
- Central Research Institute of Dentistry and Maxillofacial Surgery, 119991 Moscow, Russia
- E.V. Borovsky Institute of Dentistry, Sechenov University, 119991 Moscow, Russia
| | - Tatiana Brailovskaya
- Central Research Institute of Dentistry and Maxillofacial Surgery, 119991 Moscow, Russia
- E.V. Borovsky Institute of Dentistry, Sechenov University, 119991 Moscow, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, 119991 Moscow, Russia
| |
Collapse
|
6
|
Singh D, Memari E, He S, Yusefi H, Helfield B. Cardiac gene delivery using ultrasound: State of the field. Mol Ther Methods Clin Dev 2024; 32:101277. [PMID: 38983873 PMCID: PMC11231612 DOI: 10.1016/j.omtm.2024.101277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Over the past two decades, there has been tremendous and exciting progress toward extending the use of medical ultrasound beyond a traditional imaging tool. Ultrasound contrast agents, typically used for improved visualization of blood flow, have been explored as novel non-viral gene delivery vectors for cardiovascular therapy. Given this adaptation to ultrasound contrast-enhancing agents, this presents as an image-guided and site-specific gene delivery technique with potential for multi-gene and repeatable delivery protocols-overcoming some of the limitations of alternative gene therapy approaches. In this review, we provide an overview of the studies to date that employ this technique toward cardiac gene therapy using cardiovascular disease animal models and summarize their key findings.
Collapse
Affiliation(s)
- Davindra Singh
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Elahe Memari
- Department of Physics, Concordia University, Montreal, QC, Canada
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Hossein Yusefi
- Department of Physics, Concordia University, Montreal, QC, Canada
| | - Brandon Helfield
- Department of Biology, Concordia University, Montreal, QC, Canada
- Department of Physics, Concordia University, Montreal, QC, Canada
| |
Collapse
|
7
|
Lungu CN, Mangalagiu II, Gurau G, Mehedinti MC. Variations of VEGFR2 Chemical Space: Stimulator and Inhibitory Peptides. Int J Mol Sci 2024; 25:7787. [PMID: 39063029 PMCID: PMC11276785 DOI: 10.3390/ijms25147787] [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: 06/06/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The kinase pathway plays a crucial role in blood vessel function. Particular attention is paid to VEGFR type 2 angiogenesis and vascular morphogenesis as the tyrosine kinase pathway is preferentially activated. In silico studies were performed on several peptides that affect VEGFR2 in both stimulating and inhibitory ways. This investigation aims to examine the molecular properties of VEGFR2, a molecule primarily involved in the processes of vasculogenesis and angiogenesis. These relationships were defined by the interactions between Vascular Endothelial Growth Factor receptor 2 (VEGFR2) and the structural features of the systems. The chemical space of the inhibitory peptides and stimulators was described using topological and energetic properties. Furthermore, chimeric models of stimulating and inhibitory proteins (for VEGFR2) were computed using the protein system structures. The interaction between the chimeric proteins and VEGFR was computed. The chemical space was further characterized using complex manifolds and high-dimensional data visualization. The results show that a slightly similar chemical area is shared by VEGFR2 and stimulating and inhibitory proteins. On the other hand, the stimulator peptides and the inhibitors have distinct chemical spaces.
Collapse
Affiliation(s)
- Claudiu N. Lungu
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania; (G.G.); (M.C.M.)
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
| | - Ionel I. Mangalagiu
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
| | - Gabriela Gurau
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania; (G.G.); (M.C.M.)
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
| | - Mihaela Cezarina Mehedinti
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania; (G.G.); (M.C.M.)
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
| |
Collapse
|
8
|
Ramses R, Kennedy S, Good R, Oldroyd KG, Mcginty S. Performance of drug-coated balloons in coronary and below-the-knee arteries: Anatomical, physiological and pathological considerations. Vascul Pharmacol 2024; 155:107366. [PMID: 38479462 DOI: 10.1016/j.vph.2024.107366] [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: 09/17/2023] [Revised: 02/24/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
Below-the-knee (infrapopliteal) atherosclerotic disease, which presents as chronic limb-threatening ischemia (CLTI) in nearly 50% of patients, represents a treatment challenge when it comes to the endovascular intervention arm of management. Due to reduced tissue perfusion, patients usually experience pain at rest and atrophic changes correlated to the extent of the compromised perfusion. Unfortunately, the prognosis remains unsatisfactory with 30% of patients requiring major amputation and a mortality rate of 25% within 1 year. To date, randomized multicentre trials of endovascular intervention have shown that drug-eluting stents (DES) increase patency rate and lower target lesion revascularization rate compared to plain balloon angioplasty and bare-metal stents. The majority of these trials recruited patients with focal infrapopliteal lesions, while most patients requiring endovascular intervention have complex and diffuse atherosclerotic disease. Moreover, due to the nature of the infrapopliteal arteries, the use of long DES is limited. Following recent results of drug-coated balloons (DCBs) in the treatment of femoropopliteal and coronary arteries, it was hoped that similar effective results would be achieved in the infrapopliteal arteries. In reality, multicentre trials have failed to support the proposed hypothesis and no advantage was found in using DCBs in comparison to plain balloon angioplasty. This review aims to explore anatomical, physiological and pathological differences between lesions of the infrapopliteal and coronary arteries to explain the differences in outcome when using DCBs.
Collapse
Affiliation(s)
- Rafic Ramses
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy; Division of Biomedical Engineering, University of Glasgow, United Kingdom
| | - Simon Kennedy
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom
| | - Richard Good
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom; West of Scotland Regional Heart & Lung Centre, Golden Jubilee National Hospital, Glasgow, United Kingdom
| | - Keith G Oldroyd
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom; West of Scotland Regional Heart & Lung Centre, Golden Jubilee National Hospital, Glasgow, United Kingdom
| | - Sean Mcginty
- Division of Biomedical Engineering, University of Glasgow, United Kingdom.
| |
Collapse
|
9
|
Mullin JA, Rahmani E, Kiick KL, Sullivan MO. Growth factors and growth factor gene therapies for treating chronic wounds. Bioeng Transl Med 2024; 9:e10642. [PMID: 38818118 PMCID: PMC11135157 DOI: 10.1002/btm2.10642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 06/01/2024] Open
Abstract
Chronic wounds are an unmet clinical need affecting millions of patients globally, and current standards of care fail to consistently promote complete wound closure and prevent recurrence. Disruptions in growth factor signaling, a hallmark of chronic wounds, have led researchers to pursue growth factor therapies as potential supplements to standards of care. Initial studies delivering growth factors in protein form showed promise, with a few formulations reaching clinical trials and one obtaining clinical approval. However, protein-form growth factors are limited by instability and off-target effects. Gene therapy offers an alternative approach to deliver growth factors to the chronic wound environment, but safety concerns surrounding gene therapy as well as efficacy challenges in the gene delivery process have prevented clinical translation. Current growth factor delivery and gene therapy approaches have primarily used single growth factor formulations, but recent efforts have aimed to develop multi-growth factor approaches that are better suited to address growth factor insufficiencies in the chronic wound environment, and these strategies have demonstrated improved efficacy in preclinical studies. This review provides an overview of chronic wound healing, emphasizing the need and potential for growth factor therapies. It includes a summary of current standards of care, recent advances in growth factor, cell-based, and gene therapy approaches, and future perspectives for multi-growth factor therapeutics.
Collapse
Affiliation(s)
- James A. Mullin
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Erfan Rahmani
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Kristi L. Kiick
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
- Department of Materials Science and EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Millicent O. Sullivan
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
| |
Collapse
|
10
|
Espuche B, Moya SE, Calderón M. Nanogels: Smart tools to enlarge the therapeutic window of gene therapy. Int J Pharm 2024; 653:123864. [PMID: 38309484 DOI: 10.1016/j.ijpharm.2024.123864] [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: 10/21/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
Gene therapy can potentially treat a great number of diseases, from cancer to rare genetic disorders. Very recently, the development and emergency approval of nucleic acid-based COVID-19 vaccines confirmed its strength and versatility. However, gene therapy encounters limitations due to the lack of suitable carriers to vectorize therapeutic genetic material inside target cells. Nanogels are highly hydrated nano-size crosslinked polymeric networks that have been used in many biomedical applications, from drug delivery to tissue engineering and diagnostics. Due to their easy production, tunability, and swelling properties they have called the attention as promising vectors for gene delivery. In this review, nanogels are discussed as vectors for nucleic acid delivery aiming to enlarge gene therapy's therapeutic window. Recent works highlighting the optimization of inherent transfection efficiency and biocompatibility are reviewed here. The importance of the monomer choice, along with the internal structure, surface decoration, and responsive features are outlined for the different transfection modalities. The possible sources of toxicological endpoints in nanogels are analyzed, and the strategies to limit them are compared. Finally, perspectives are discussed to identify the remining challenges for the nanogels before their translation to the market as transfection agents.
Collapse
Affiliation(s)
- Bruno Espuche
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain; POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain
| | - Sergio E Moya
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain.
| |
Collapse
|
11
|
Zhu Y, Liu R, Zhao X, Kang C, Yang D, Ge G. VEGF overexpression in transplanted NSCs promote recovery of neurological function in rats with cerebral ischemia by modulating the Wnt signal transduction pathway. Neurosci Lett 2024; 824:137668. [PMID: 38331020 DOI: 10.1016/j.neulet.2024.137668] [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: 10/18/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Neural stem cell transplantation is a good method to treat stroke, but the mechanism is still unclear. Therefore, this study aims to explore the regulatory mechanism of VEGF overexpression in transplanted NSCs to promote the recovery of neural function in ischemic rats by regulating Wnt signal transduction pathways. We amplified VEGF gene fragments by PCR and transfected them into NSCs with Ad5 adenovirus. Rat brain IRI model was established by MCAO method, and VEGF transfected NSCs (VEGF-NSCs) were transplanted 24 h after successful IRI model. One week after the transplant, cognitive function was assessed using a neurological deficit score; Brain injury was assessed by histopathology; Photochemical and ELISA methods were used to detect oxidative stress markers and inflammatory factors, respectively. Western blotting has been detected in molecules of the Wnt signaling pathway. The results showed that the transduced NSCs express VEGF at least for 14 days. VEGF-NSCs transplantation (VNT) improved spatial learning and memory in rats, and inhibited oxidative stress injury, inflammatory response, and histopathological injury. VNT also resulted in significant changes in the phosphorylation levels of β-catenin and GSK-3β proteins, ultimately triggering activation of the Wnt signal transduction pathway. These results suggest that the neuroprotective effects of VNT may be related to the regulation of the Wnt signal transduction pathway.
Collapse
Affiliation(s)
- Yizhen Zhu
- Class 3, Nursing, Grade 2023, Guizhou Medical University, Guizhou 551113, PR China.
| | - Ruojing Liu
- Department of Human Anatomy, Guizhou Medical University, Guizhou 561113, PR China.
| | - Xue Zhao
- Department of Human Anatomy, Guizhou Medical University, Guizhou 561113, PR China.
| | - Chaosheng Kang
- Department of Human Anatomy, Guizhou Medical University, Guizhou 561113, PR China.
| | - Dan Yang
- Department of Human Anatomy, Guizhou Medical University, Guizhou 561113, PR China.
| | - Guo Ge
- Department of Human Anatomy, Guizhou Medical University, Guizhou 561113, PR China.
| |
Collapse
|
12
|
He Q, Wang Y, Fang C, Feng Z, Yin M, Huang J, Ma Y, Mo Z. Advancing stroke therapy: A deep dive into early phase of ischemic stroke and recanalization. CNS Neurosci Ther 2024; 30:e14634. [PMID: 38379112 PMCID: PMC10879038 DOI: 10.1111/cns.14634] [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: 11/27/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Ischemic stroke, accounting for the majority of stroke events, significantly contributes to global morbidity and mortality. Vascular recanalization therapies, namely intravenous thrombolysis and mechanical thrombectomy, have emerged as critical interventions, yet their success hinges on timely application and patient-specific factors. This review focuses on the early phase pathophysiological mechanisms of ischemic stroke and the nuances of recanalization. It highlights the dual role of neutrophils in tissue damage and repair, and the critical involvement of the blood-brain barrier (BBB) in stroke outcomes. Special emphasis is placed on ischemia-reperfusion injury, characterized by oxidative stress, inflammation, and endothelial dysfunction, which paradoxically exacerbates cerebral damage post-revascularization. The review also explores the potential of targeting molecular pathways involved in BBB integrity and inflammation to enhance the efficacy of recanalization therapies. By synthesizing current research, this paper aims to provide insights into optimizing treatment protocols and developing adjuvant neuroprotective strategies, thereby advancing stroke therapy and improving patient outcomes.
Collapse
Affiliation(s)
- Qianyan He
- Department of Neurology, Stroke CenterThe First Hospital of Jilin UniversityJilinChina
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Yueqing Wang
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Cheng Fang
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Ziying Feng
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Meifang Yin
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Juyang Huang
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yinzhong Ma
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine HospitalThe Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhenGuangdongChina
| |
Collapse
|
13
|
Tung LW, Groppa E, Soliman H, Lin B, Chang C, Cheung CW, Ritso M, Guo D, Rempel L, Sinha S, Eisner C, Brassard J, McNagny K, Biernaskie J, Rossi F. Spatiotemporal signaling underlies progressive vascular rarefaction in myocardial infarction. Nat Commun 2023; 14:8498. [PMID: 38129410 PMCID: PMC10739910 DOI: 10.1038/s41467-023-44227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Therapeutic angiogenesis represents a promising avenue to revascularize the ischemic heart. Its limited success is partly due to our poor understanding of the cardiac stroma, specifically mural cells, and their response to ischemic injury. Here, we combine single-cell and positional transcriptomics to assess the behavior of mural cells within the healing heart. In response to myocardial infarction, mural cells adopt an altered state closely associated with the infarct and retain a distinct lineage from fibroblasts. This response is concurrent with vascular rarefaction and reduced vascular coverage by mural cells. Positional transcriptomics reveals that the infarcted heart is governed by regional-dependent and temporally regulated programs. While the remote zone acts as an important source of pro-angiogenic signals, the infarct zone is accentuated by chronic activation of anti-angiogenic, pro-fibrotic, and inflammatory cues. Together, our work unveils the spatiotemporal programs underlying cardiac repair and establishes an association between vascular deterioration and mural cell dysfunction.
Collapse
Affiliation(s)
- Lin Wei Tung
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Elena Groppa
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Borea Therapeutics, Scuola Internazionale Superiore di Studi Avanzati, Via Bonomea, 265, 34136, Trieste, Italy
| | - Hesham Soliman
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Aspect Biosystems, 1781 W 75th Ave, Vancouver, BC, V6P 6P2, Canada
- Faculty of Pharmaceutical Sciences, Minia University, Minia, Egypt
| | - Bruce Lin
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Chihkai Chang
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Chun Wai Cheung
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Morten Ritso
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - David Guo
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lucas Rempel
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Sarthak Sinha
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christine Eisner
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Julyanne Brassard
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Kelly McNagny
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Fabio Rossi
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
14
|
Guo L, Yang Q, Wei R, Zhang W, Yin N, Chen Y, Xu C, Li C, Carney RP, Li Y, Feng M. Enhanced pericyte-endothelial interactions through NO-boosted extracellular vesicles drive revascularization in a mouse model of ischemic injury. Nat Commun 2023; 14:7334. [PMID: 37957174 PMCID: PMC10643472 DOI: 10.1038/s41467-023-43153-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Despite improvements in medical and surgical therapies, a significant portion of patients with critical limb ischemia (CLI) are considered as "no option" for revascularization. In this work, a nitric oxide (NO)-boosted and activated nanovesicle regeneration kit (n-BANK) is constructed by decorating stem cell-derived nanoscale extracellular vesicles with NO nanocages. Our results demonstrate that n-BANKs could store NO in endothelial cells for subsequent release upon pericyte recruitment for CLI revascularization. Notably, n-BANKs enable endothelial cells to trigger eNOS activation and form tube-like structures. Subsequently, eNOS-derived NO robustly recruits pericytes to invest nascent endothelial cell tubes, giving rise to mature blood vessels. Consequently, n-BANKs confer complete revascularization in female mice following CLI, and thereby achieve limb preservation and restore the motor function. In light of n-BANK evoking pericyte-endothelial interactions to create functional vascular networks, it features promising therapeutic potential in revascularization to reduce CLI-related amputations, which potentially impact regeneration medicine.
Collapse
Affiliation(s)
- Ling Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China.
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, P. R. China.
| | - Qiang Yang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Runxiu Wei
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Wenjun Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Na Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Yuling Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Chao Xu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, P. R. China
| | - Changrui Li
- Guangzhou Zhixin High School, Zhixin South Road, Guangzhou, 510080, P.R. China
| | - Randy P Carney
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, USA.
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, 95616, USA.
| | - Min Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China.
| |
Collapse
|
15
|
Chastagnier L, Marquette C, Petiot E. In situ transient transfection of 3D cell cultures and tissues, a promising tool for tissue engineering and gene therapy. Biotechnol Adv 2023; 68:108211. [PMID: 37463610 DOI: 10.1016/j.biotechadv.2023.108211] [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: 12/15/2022] [Revised: 04/26/2023] [Accepted: 07/09/2023] [Indexed: 07/20/2023]
Abstract
Various research fields use the transfection of mammalian cells with genetic material to induce the expression of a target transgene or gene silencing. It is a tool widely used in biological research, bioproduction, and therapy. Current transfection protocols are usually performed on 2D adherent cells or suspension cultures. The important rise of new gene therapies and regenerative medicine in the last decade raises the need for new tools to empower the in situ transfection of tissues and 3D cell cultures. This review will present novel in situ transfection methods based on a chemical or physical non-viral transfection of cells in tissues and 3D cultures, discuss the advantages and remaining gaps, and propose future developments and applications.
Collapse
Affiliation(s)
- Laura Chastagnier
- 3D Innovation Lab - 3d.FAB - ICBMS, University Claude Bernard Lyon 1, Université Lyon 1, CNRS, INSA, CPE-Lyon, UMR 5246, bat. Lederer, 5 rue Gaston Berger, 69100 Villeurbanne, France
| | - Christophe Marquette
- 3D Innovation Lab - 3d.FAB - ICBMS, University Claude Bernard Lyon 1, Université Lyon 1, CNRS, INSA, CPE-Lyon, UMR 5246, bat. Lederer, 5 rue Gaston Berger, 69100 Villeurbanne, France
| | - Emma Petiot
- 3D Innovation Lab - 3d.FAB - ICBMS, University Claude Bernard Lyon 1, Université Lyon 1, CNRS, INSA, CPE-Lyon, UMR 5246, bat. Lederer, 5 rue Gaston Berger, 69100 Villeurbanne, France.
| |
Collapse
|
16
|
Chen C, Wang J, Liu C, Hu J, Liu L. Pioneering therapies for post-infarction angiogenesis: Insight into molecular mechanisms and preclinical studies. Biomed Pharmacother 2023; 166:115306. [PMID: 37572633 DOI: 10.1016/j.biopha.2023.115306] [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: 06/16/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
Acute myocardial infarction (MI), despite significant progress in its treatment, remains a leading cause of chronic heart failure and cardiovascular events such as cardiac arrest. Promoting angiogenesis in the myocardial tissue after MI to restore blood flow in the ischemic and hypoxic tissue is considered an effective treatment strategy. The repair of the myocardial tissue post-MI involves a robust angiogenic response, with mechanisms involved including endothelial cell proliferation and migration, capillary growth, changes in the extracellular matrix, and stabilization of pericytes for neovascularization. In this review, we provide a detailed overview of six key pathways in angiogenesis post-MI: the PI3K/Akt/mTOR signaling pathway, the Notch signaling pathway, the Wnt/β-catenin signaling pathway, the Hippo signaling pathway, the Sonic Hedgehog signaling pathway, and the JAK/STAT signaling pathway. We also discuss novel therapeutic approaches targeting these pathways, including drug therapy, gene therapy, protein therapy, cell therapy, and extracellular vesicle therapy. A comprehensive understanding of these key pathways and their targeted therapies will aid in our understanding of the pathological and physiological mechanisms of angiogenesis after MI and the development and application of new treatment strategies.
Collapse
Affiliation(s)
- Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Chao Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| |
Collapse
|
17
|
Zhao L, Lee AS, Sasagawa K, Sokol J, Wang Y, Ransom RC, Zhao X, Ma C, Steininger HM, Koepke LS, Borrelli MR, Brewer RE, Lee LL, Huang X, Ambrosi TH, Sinha R, Hoover MY, Seita J, Weissman IL, Wu JC, Wan DC, Xiao J, Longaker MT, Nguyen PK, Chan CK. A Combination of Distinct Vascular Stem/Progenitor Cells for Neovascularization and Ischemic Rescue. Arterioscler Thromb Vasc Biol 2023; 43:1262-1277. [PMID: 37051932 PMCID: PMC10281192 DOI: 10.1161/atvbaha.122.317943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/09/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND Peripheral vascular disease remains a leading cause of vascular morbidity and mortality worldwide despite advances in medical and surgical therapy. Besides traditional approaches, which can only restore blood flow to native arteries, an alternative approach is to enhance the growth of new vessels, thereby facilitating the physiological response to ischemia. METHODS The ActinCreER/R26VT2/GK3 Rainbow reporter mouse was used for unbiased in vivo survey of injury-responsive vasculogenic clonal formation. Prospective isolation and transplantation were used to determine vessel-forming capacity of different populations. Single-cell RNA-sequencing was used to characterize distinct vessel-forming populations and their interactions. RESULTS Two populations of distinct vascular stem/progenitor cells (VSPCs) were identified from adipose-derived mesenchymal stromal cells: VSPC1 is CD45-Ter119-Tie2+PDGFRa-CD31+CD105highSca1low, which gives rise to stunted vessels (incomplete tubular structures) in a transplant setting, and VSPC2 which is CD45-Ter119-Tie2+PDGFRa+CD31-CD105lowSca1high and forms stunted vessels and fat. Interestingly, cotransplantation of VSPC1 and VSPC2 is required to form functional vessels that improve perfusion in the mouse hindlimb ischemia model. Similarly, VSPC1 and VSPC2 populations isolated from human adipose tissue could rescue the ischemic condition in mice. CONCLUSIONS These findings suggest that autologous cotransplantation of synergistic VSPCs from nonessential adipose tissue can promote neovascularization and represents a promising treatment for ischemic disease.
Collapse
Affiliation(s)
- Liming Zhao
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
| | - Andrew S. Lee
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, China (A.S.L.)
- Institute for Cancer Research, Shenzhen Bay Laboratory, China (A.S.L.)
| | - Koki Sasagawa
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Jan Sokol
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
- Center for Integrative Medical Sciences and Advanced Data Science Project, RIKEN, Tokyo, Japan (J.S.)
| | - Yuting Wang
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
| | - Ryan C. Ransom
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Xin Zhao
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Chao Ma
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Holly M. Steininger
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Lauren S. Koepke
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Mimi R. Borrelli
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Rachel E. Brewer
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Lorene L.Y. Lee
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Xianxi Huang
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Thomas H. Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Malachia Y. Hoover
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Jun Seita
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
- Department of Developmental Biology (I.L.W., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, China (A.S.L.)
- Institute for Cancer Research, Shenzhen Bay Laboratory, China (A.S.L.)
- Center for Integrative Medical Sciences and Advanced Data Science Project, RIKEN, Tokyo, Japan (J.S.)
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Developmental Biology (I.L.W., C.K.F.C.), Stanford University School of Medicine, CA
| | - Joseph C. Wu
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Derrick C. Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
| | - Michael T. Longaker
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Patricia K. Nguyen
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Charles K.F. Chan
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Developmental Biology (I.L.W., C.K.F.C.), Stanford University School of Medicine, CA
| |
Collapse
|
18
|
Oh GC, Choi YJ, Park BW, Ban K, Park HJ. Are There Hopeful Therapeutic Strategies to Regenerate the Infarcted Hearts? Korean Circ J 2023; 53:367-386. [PMID: 37271744 DOI: 10.4070/kcj.2023.0098] [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: 04/10/2023] [Accepted: 04/26/2023] [Indexed: 06/06/2023] Open
Abstract
Ischemic heart disease remains the primary cause of morbidity and mortality worldwide. Despite significant advancements in pharmacological and revascularization techniques in the late 20th century, heart failure prevalence after myocardial infarction has gradually increased over the last 2 decades. After ischemic injury, pathological remodeling results in cardiomyocytes (CMs) loss and fibrosis, which leads to impaired heart function. Unfortunately, there are no clinical therapies to regenerate CMs to date, and the adult heart's limited turnover rate of CMs hinders its ability to self-regenerate. In this review, we present novel therapeutic strategies to regenerate injured myocardium, including (1) reconstruction of cardiac niche microenvironment, (2) recruitment of functional CMs by promoting their proliferation or differentiation, and (3) organizing 3-dimensional tissue construct beyond the CMs. Additionally, we highlight recent mechanistic insights that govern these strategies and identify current challenges in translating these approaches to human patients.
Collapse
Affiliation(s)
- Gyu-Chul Oh
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Yeon-Jik Choi
- Division of Cardiology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Bong-Woo Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong.
| | - Hun-Jun Park
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
19
|
Shams S, Stilhano RS, Silva EA. Harnessing EGLN1 Gene Editing to Amplify HIF-1α and Enhance Human Angiogenic Response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.542734. [PMID: 37398294 PMCID: PMC10312464 DOI: 10.1101/2023.05.29.542734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Therapeutic angiogenesis has been the focus of hundreds of clinical trials but approval for human treatment remains elusive. Current strategies often rely on the upregulation of a single proangiogenic factor, which fails to recapitulate the complex response needed in hypoxic tissues. Hypoxic oxygen tensions dramatically decrease the activity of hypoxia inducible factor prolyl hydroxylase 2 (PHD2), the primary oxygen sensing portion of the hypoxia inducible factor 1 alpha (HIF-1α) proangiogenic master regulatory pathway. Repressing PHD2 activity increases intracellular levels of HIF-1α and impacts the expression of hundreds of downstream genes directly associated with angiogenesis, cell survival, and tissue homeostasis. This study explores activating the HIF-1α pathway through Sp Cas9 knockout of the PHD2 encoding gene EGLN1 as an innovative in situ therapeutic angiogenesis strategy for chronic vascular diseases. Our findings demonstrate that even low editing rates of EGLN1 lead to a strong proangiogenic response regarding proangiogenic gene transcription, protein production, and protein secretion. In addition, we show that secreted factors of EGLN1 edited cell cultures may enhance human endothelial cell neovascularization activity in the context of proliferation and motility. Altogether, this study reveals that EGLN1 gene editing shows promise as a potential therapeutic angiogenesis strategy.
Collapse
|
20
|
Brlecic PE, Bonham CA, Rosengart TK, Mathison M. Direct cardiac reprogramming: A new technology for cardiac repair. J Mol Cell Cardiol 2023; 178:51-58. [PMID: 36965701 PMCID: PMC10124164 DOI: 10.1016/j.yjmcc.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/03/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Cardiovascular disease is one of the leading causes of morbidity and mortality worldwide, with myocardial infarctions being amongst the deadliest manifestations. Reduced blood flow to the heart can result in the death of cardiac tissue, leaving affected patients susceptible to further complications and recurrent disease. Further, contemporary management typically involves a pharmacopeia to manage the metabolic conditions contributing to atherosclerotic and hypertensive heart disease, rather than regeneration of the damaged myocardium. With modern healthcare extending lifespan, a larger demographic will be at risk for heart disease, driving the need for novel therapeutics that surpass those currently available in efficacy. Transdifferentiation and cellular reprogramming have been looked to as potential methods for the treatment of diseases throughout the body. Specifically targeting the fibrotic cells in cardiac scar tissue as a source to be reprogrammed into induced cardiomyocytes remains an appealing option. This review aims to highlight the history of and advances in cardiac reprogramming and describe its translational potential as a treatment for cardiovascular disease.
Collapse
Affiliation(s)
- Paige E Brlecic
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Clark A Bonham
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Todd K Rosengart
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
21
|
White AL, Bix GJ. VEGFA Isoforms as Pro-Angiogenic Therapeutics for Cerebrovascular Diseases. Biomolecules 2023; 13:biom13040702. [PMID: 37189449 DOI: 10.3390/biom13040702] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Therapeutic angiogenesis has long been considered a viable treatment for vasculature disruptions, including cerebral vasculature diseases. One widely-discussed treatment method to increase angiogenesis is vascular endothelial growth factor (VEGF) A. In animal models, treatment with VEGFA proved beneficial, resulting in increased angiogenesis, increased neuronal density, and improved outcome. However, VEGFA administration in clinical trials has thus far failed to replicate the promising results seen in animal models. The lack of beneficial effects in humans and the difficulty in medicinal translation may be due in part to administration methods and VEGFA's ability to increase vascular permeability. One solution to mitigate the side effects of VEGFA may be found in the VEGFA isoforms. VEGFA is able to produce several different isoforms through alternative splicing. Each VEGFA isoform interacts differently with both the cellular components and the VEGF receptors. Because of the different biological effects elicited, VEGFA isoforms may hold promise as a tangible potential therapeutic for cerebrovascular diseases.
Collapse
Affiliation(s)
- Amanda Louise White
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Gregory Jaye Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70122, USA
| |
Collapse
|
22
|
Grosso A, Lunger A, Burger MG, Briquez PS, Mai F, Hubbell JA, Schaefer DJ, Banfi A, Di Maggio N. VEGF dose controls the coupling of angiogenesis and osteogenesis in engineered bone. NPJ Regen Med 2023; 8:15. [PMID: 36914692 PMCID: PMC10011536 DOI: 10.1038/s41536-023-00288-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/23/2023] [Indexed: 03/16/2023] Open
Abstract
Vascular endothelial growth factor-A (VEGF) physiologically regulates both angiogenesis and osteogenesis, but its application in bone tissue engineering led to contradictory outcomes. A poorly understood aspect is how VEGF dose impacts the coordination between these two processes. Taking advantage of a unique and highly tunable platform, here we dissected the effects of VEGF dose over a 1,000-fold range in the context of tissue-engineered osteogenic grafts. We found that osteo-angiogenic coupling is exquisitely dependent on VEGF dose and that only a tightly defined dose range could stimulate both vascular invasion and osteogenic commitment of progenitors, with significant improvement in bone formation. Further, VEGF dose regulated Notch1 activation and the induction of a specific pro-osteogenic endothelial phenotype, independently of the promotion of vascular invasion. Therefore, in a therapeutic perspective, fine-tuning of VEGF dose in the signaling microenvironment is key to ensure physiological coupling of accelerated vascular invasion and improved bone formation.
Collapse
Affiliation(s)
- Andrea Grosso
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Alexander Lunger
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.,Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Petersgraben 4, 4031, Basel, Switzerland
| | - Maximilian G Burger
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.,Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Petersgraben 4, 4031, Basel, Switzerland
| | - Priscilla S Briquez
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S Ellis Ave, Chicago, IL, 60637, USA.,Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Francesca Mai
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S Ellis Ave, Chicago, IL, 60637, USA
| | - Dirk J Schaefer
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.,Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Petersgraben 4, 4031, Basel, Switzerland
| | - Andrea Banfi
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland. .,Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Petersgraben 4, 4031, Basel, Switzerland.
| | - Nunzia Di Maggio
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.
| |
Collapse
|
23
|
Everts PA, Panero AJ. Basic Science of Autologous Orthobiologics. Phys Med Rehabil Clin N Am 2023; 34:25-47. [DOI: 10.1016/j.pmr.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
24
|
Cao W, Zhang H, Zhou N, Zhou R, Zhang X, Yin J, Deng J, Ao X, Shi C. Functional recovery of myocardial infarction by specific EBP-PR1P peptides bridging injectable cardiac extracellular matrix and vascular endothelial growth factor. J Biomed Mater Res A 2022; 111:995-1005. [PMID: 36579729 DOI: 10.1002/jbm.a.37483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/26/2022] [Accepted: 12/08/2022] [Indexed: 12/30/2022]
Abstract
Vascular endothelial growth factor (VEGF) is the most potent angiogenic factor and plays an important role in therapy of myocardial infarction (MI). Currently, how to retain regional concentration and decrease rapid diffusion is critical for its clinical application of VEGF. In recent years, the application of targeting peptides has been developed rapidly and provides new strategies for the sustained release of VEGF. In present study, a bi-functional EBP-PR1P peptide was designed and bridged VEGF to injectable cardiac extracellular matrix (c-ECM). Through EBP-PR1P peptides, VEGF could specifically bind with c-ECM to realize the sustained release, without impacting the bioactivity of VEGF. Then VEGF/EBP-PR1P/c-ECM scaffolds were constructed and administrated into rats with MI. The results showed VEGF/EBP-PR1P/c-ECM could promote angiogenesis, protect cardiomyocytes survival against apoptosis, and improve the recovery of cardiac function. In addition, the mechanism of EBP-PR1P/VEGF was also investigated which canonical downstream of VEGF-Akt signaling pathway was activated. These results showed specific VEGF/EBP-PR1P/c-ECM scaffolds served as promising delivery system for VEGF that facilitated the functional recovery of MI.
Collapse
Affiliation(s)
- Wenxuan Cao
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Hong Zhang
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Ning Zhou
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Runxue Zhou
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaojing Zhang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jia Yin
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jin Deng
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xiang Ao
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Chunying Shi
- School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
25
|
Pajula J, Lähteenvuo J, Lähteenvuo M, Honkonen K, Halonen P, Hätinen OP, Kuivanen A, Heikkilä M, Nurro J, Hartikainen J, Ylä-Herttuala S. Adenoviral VEGF-D ΔN ΔC gene therapy for myocardial ischemia. Front Bioeng Biotechnol 2022; 10:999226. [PMID: 36619378 PMCID: PMC9817830 DOI: 10.3389/fbioe.2022.999226] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Cardiovascular diseases are the leading cause of death globally. In spite of the availability of improved treatments, there is still a large group of chronic ischemia patients who suffer from significant symptoms and disability. Thus, there is a clear need to develop new treatment strategies for these patients. Therapeutic angiogenesis is a novel therapy method which has shown promising results in preclinical studies. In this study, we evaluated safety and efficacy of adenoviral (Ad) VEGF-DΔNΔC gene transfer for the treatment of myocardial ischemia in a pig model. Methods: Adenoviral VEGF-DΔNΔC gene transfer was given to pigs (n = 26) via intramyocardial injections using an electromechanical injection catheter. Angiogenic effects were evaluated in an acute myocardial infarction model (n = 18) and functionality of the lymphatic vessels were tested in healthy porcine myocardium (n = 8). AdLacZ was used as a control. Results: AdVEGF-DΔNΔC induced safe and effective myocardial angiogenesis by inducing a four-fold increase in mean capillary area at the edge of the myocardial infarct six days after the gene transfer relative to the control AdLacZ group. The effect was sustained over 21 days after the gene transfer, and there were no signs of vessels regression. AdVEGF-DΔNΔC also increased perfusion 3.4-fold near the infarct border zone relative to the control as measured by fluorescent microspheres. Ejection fraction was 8.7% higher in the AdVEGF-DΔNΔC treated group 21 days after the gene transfer relative to the AdLacZ control group. Modified Miles assay detected a transient increase in plasma protein extravasation after the AdVEGF-DΔNΔC treatment and a mild accumulation of pericardial effusate was observed at d6. However, AdVEGF-DΔNΔC also induced the growth of functional lymphatic vasculature, and the amount of pericardial fluid and level of vascular permeability had returned to normal by d21. Conclusion: Endovascular intramyocardial AdVEGF-DΔNΔC gene therapy proved to be safe and effective in the acute porcine myocardial infarction model and provides a new potential treatment option for patients with severe coronary heart disease.
Collapse
Affiliation(s)
- Juho Pajula
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | | | - Markku Lähteenvuo
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Krista Honkonen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Paavo Halonen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | | | - Antti Kuivanen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Minja Heikkilä
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Jussi Nurro
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Juha Hartikainen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland,Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland,Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland,*Correspondence: Seppo Ylä-Herttuala,
| |
Collapse
|
26
|
Wasko R, Bridges K, Pannone R, Sidhu I, Xing Y, Naik S, Miller-Jensen K, Horsley V. Langerhans cells are essential components of the angiogenic niche during murine skin repair. Dev Cell 2022; 57:2699-2713.e5. [PMID: 36493773 PMCID: PMC10848275 DOI: 10.1016/j.devcel.2022.11.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/28/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022]
Abstract
Angiogenesis, the growth of new blood vessels from pre-existing vessels, occurs during development, injury repair, and tumorigenesis to deliver oxygen, immune cells, and nutrients to tissues. Defects in angiogenesis occur in cardiovascular and inflammatory diseases, and chronic, non-healing wounds, yet treatment options are limited. Here, we provide a map of the early angiogenic niche by analyzing single-cell RNA sequencing of mouse skin wound healing. Our data implicate Langerhans cells (LCs), phagocytic, skin-resident immune cells, in driving angiogenesis during skin repair. Using lineage-driven reportersw, three-dimensional (3D) microscopy, and mouse genetics, we show that LCs are situated at the endothelial cell leading edge in mouse skin wounds and are necessary for angiogenesis during repair. These data provide additional future avenues for the control of angiogenesis to treat disease and chronic wounds and extend the function of LCs beyond their canonical role in antigen presentation and T cell immunity.
Collapse
Affiliation(s)
- Renee Wasko
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Kate Bridges
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Rebecca Pannone
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Ikjot Sidhu
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Yue Xing
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Shruti Naik
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Kathryn Miller-Jensen
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| | - Valerie Horsley
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
27
|
Extracellular Matrix-Based Approaches in Cardiac Regeneration: Challenges and Opportunities. Int J Mol Sci 2022; 23:ijms232415783. [PMID: 36555424 PMCID: PMC9779713 DOI: 10.3390/ijms232415783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiac development is characterized by the active proliferation of different cardiac cell types, in particular cardiomyocytes and endothelial cells, that eventually build the beating heart. In mammals, these cells lose their regenerative potential early after birth, representing a major obstacle to our current capacity to restore the myocardial structure and function after an injury. Increasing evidence indicates that the cardiac extracellular matrix (ECM) actively regulates and orchestrates the proliferation, differentiation, and migration of cardiac cells within the heart, and that any change in either the composition of the ECM or its mechanical properties ultimately affect the behavior of these cells throughout one's life. Thus, understanding the role of ECMs' proteins and related signaling pathways on cardiac cell proliferation is essential to develop effective strategies fostering the regeneration of a damaged heart. This review provides an overview of the components of the ECM and its mechanical properties, whose function in cardiac regeneration has been elucidated, with a major focus on the strengths and weaknesses of the experimental models so far exploited to demonstrate the actual pro-regenerative capacity of the components of the ECM and to translate this knowledge into new therapies.
Collapse
|
28
|
Jiang Q, Ma Y, Zhao Y, Yao MD, Zhu Y, Zhang QY, Yan B. tRNA-derived fragment tRF-1001: A novel anti-angiogenic factor in pathological ocular angiogenesis. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 30:407-420. [DOI: 10.1016/j.omtn.2022.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022]
|
29
|
Peng H, He X, Wang Q. Targeted drug delivery system for ovarian cancer microenvironment: Improving the effects of immunotherapy. Front Immunol 2022; 13:1035997. [PMID: 36405688 PMCID: PMC9670735 DOI: 10.3389/fimmu.2022.1035997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Immunotherapies have shown modest benefits in the current clinical trials for ovarian cancer. The tumor microenvironment (TME) in an immunosuppressive phenotype contributes to this “failure” of immunotherapy in ovarian cancer. Many stromal cell types in the TME (e.g., tumor-associated macrophages and fibroblasts) have been identified as having plasticity in pro- and antitumor activities and are responsible for suppressing the antitumor immune response. Thus, the TME is an extremely valuable target for adjuvant interventions to improve the effects of immunotherapy. The current strategies targeting the TME include: 1) eliminating immunosuppressive cells or transforming them into immunostimulatory phenotypes and 2) inhibiting their immunosuppressive or pro-tumor production. Most of the effective agents used in the above strategies are genetic materials (e.g., cDNA, mRNA, or miRNA), proteins, or other small molecules (e.g., peptides), which are limited in their target and instability. Various formulations of drug delivery system (DDS) have been designed to realize the controlled release and targeting delivery of these agents to the tumor sites. Nanoparticles and liposomes are the most frequently exploited materials. Based on current evidence from preclinical and clinical studies, the future of the DDS is promising in cancer immunotherapy since the combination of agents with a DDS has shown increased efficacy and decreased toxicities compared with free agents. In the future, more efforts are needed to further identify the hallmarks and biomarkers in the ovarian TME, which is crucial for the development of more effective, safe, and personalized DDSs.
Collapse
|
30
|
Unlocking the promise of mRNA therapeutics. Nat Biotechnol 2022; 40:1586-1600. [PMID: 36329321 DOI: 10.1038/s41587-022-01491-z] [Citation(s) in RCA: 189] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/11/2022] [Accepted: 07/07/2022] [Indexed: 11/06/2022]
Abstract
The extraordinary success of mRNA vaccines against coronavirus disease 2019 (COVID-19) has renewed interest in mRNA as a means of delivering therapeutic proteins. Early clinical trials of mRNA therapeutics include studies of paracrine vascular endothelial growth factor (VEGF) mRNA for heart failure and of CRISPR-Cas9 mRNA for a congenital liver-specific storage disease. However, a series of challenges remains to be addressed before mRNA can be established as a general therapeutic modality with broad relevance to both rare and common diseases. An array of new technologies is being developed to surmount these challenges, including approaches to optimize mRNA cargos, lipid carriers with inherent tissue tropism and in vivo percutaneous delivery systems. The judicious integration of these advances may unlock the promise of biologically targeted mRNA therapeutics, beyond vaccines and other immunostimulatory agents, for the treatment of diverse clinical indications.
Collapse
|
31
|
Wolf KG, Crawford EB, Wartan NM, Schneiderman SK, Riehl VE, Dambaeva SV, Beaman KD. Ephrin-B2-expressing natural killer cells induce angiogenesis. JVS Vasc Sci 2022; 3:336-344. [DOI: 10.1016/j.jvssci.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/11/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
|
32
|
Kujawa M, O’Meara M, Li H, Xu L, Meda Venkata SP, Nguyen H, Minjares M, Zhang K, Wang JM. MicroRNA-466 and microRNA-200 increase endothelial permeability in hyperglycemia by targeting Claudin-5. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:259-271. [PMID: 35892090 PMCID: PMC9307898 DOI: 10.1016/j.omtn.2022.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 07/04/2022] [Indexed: 01/05/2023]
Abstract
Endothelial cell (EC) permeability is essential to vascular homeostasis in diabetes. MicroRNAs are critical gene regulators whose roles in the EC permeability have yet to be characterized. This study aims to examine the change in cell permeability induced by miR-200 and miR-466 in ECs. Human aortic ECs and dermal microvascular ECs from healthy subjects and type 2 diabetic patients were used. Our in vitro experiments unveiled higher expressions of miR-200 family members and miR-466 in diabetic ECs and in healthy ECs when exposed to high glucose. Overexpression of both miR-200 and miR-466 significantly increased EC permeability through transcriptional suppression of Claudin-5, the cell tight junction protein, by directly binding to its 3' untranslated region. In a mouse model of chronic hyperglycemia mimicking type 2 diabetes in humans (db/db mice), the delayed closure rate of a full-thickness excisional wound was partly rescued by topical application of the miR-200 inhibitor. The topical application of both miR-200 and miR-466 inhibitors exhibited improved efficacy in accelerating wound closure compared with the topical application of miR-200 inhibitor alone. Our study demonstrated the potentially effective approach of miR-200/miR-466 cocktail inhibition to restore vascular integrity and tissue repair in hyperglycemia.
Collapse
Affiliation(s)
- Marisa Kujawa
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Megan O’Meara
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Hainan Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Liping Xu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Sai Pranathi Meda Venkata
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Huong Nguyen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Morgan Minjares
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Kezhong Zhang
- Centers for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
- Centers for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, USA
| |
Collapse
|
33
|
Zhang S, Tuk B, van de Peppel J, Kremers GJ, Koedam M, Pesch GR, Rahman Z, Hoogenboezem RM, Bindels EMJ, van Neck JW, Boukany PE, van Leeuwen JPTM, van der Eerden BCJ. Microfluidic evidence of synergistic effects between mesenchymal stromal cell-derived biochemical factors and biomechanical forces to control endothelial cell function. Acta Biomater 2022; 151:346-359. [PMID: 35995408 DOI: 10.1016/j.actbio.2022.08.025] [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: 03/28/2022] [Revised: 07/20/2022] [Accepted: 08/12/2022] [Indexed: 11/01/2022]
Abstract
A functional vascular system is a prerequisite for bone repair as disturbed angiogenesis often causes non-union. Paracrine factors released from human bone marrow derived mesenchymal stromal cells (BMSCs) have angiogenic effects on endothelial cells. However, whether these paracrine factors participate in blood flow dynamics within bone capillaries remains poorly understood. Here, we used two different microfluidic designs to investigate critical steps during angiogenesis and found pronounced effects of endothelial cell proliferation as well as chemotactic and mechanotactic migration induced by BMSC conditioned medium (CM). The application of BMSC-CM in dynamic cultures demonstrates that bioactive factors in combination with fluidic flow-induced biomechanical signals significantly enhanced endothelial cell migration. Transcriptional analyses of endothelial cells demonstrate the induction of a unique gene expression profile related to tricarboxylic acid cycle and energy metabolism by the combination of BMSC-CM factors and shear stress, which opens an interesting avenue to explore during fracture healing. Our results stress the importance of in vivo - like microenvironments simultaneously including biochemical, biomechanical and oxygen levels when investigating key events during vessel repair. STATEMENT OF SIGNIFICANCE: Our results demonstrate the importance of recapitulating in vivo - like microenvironments when investigating key events during vessel repair. Endothelial cells exhibit enhanced angiogenesis characteristics when simultaneous exposing them to hMSC-CM, mechanical forces and biochemical signals simultaneously. The improved angiogenesis may not only result from the direct effect of growth factors, but also by reprogramming of endothelial cell metabolism. Moreover, with this model we demonstrated a synergistic impact of mechanical forces and biochemical factors on endothelial cell behavior and the expression of genes involved in the TCA cycle and energy metabolism, which opens an interesting new avenue to stimulate angiogenesis during fracture healing.
Collapse
Affiliation(s)
- Shuang Zhang
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bastiaan Tuk
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Jeroen van de Peppel
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Gert-Jan Kremers
- Erasmus Optical Imaging Center, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Marijke Koedam
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Georg R Pesch
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Remco M Hoogenboezem
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Eric M J Bindels
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Johan W van Neck
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Johannes P T M van Leeuwen
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bram C J van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands.
| |
Collapse
|
34
|
Isoda R, Morita I, Isida A, Mikami Y, Monobe Y, Sato Y, Moriya T. Pathological Study on the Expression of Vasohibins in Peripheral Artery Disease. TOHOKU J EXP MED 2022; 258:121-128. [PMID: 35922907 DOI: 10.1620/tjem.2022.j063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Ryutaro Isoda
- Department of Pathology, Kawasaki Medical School.,Department of Surgery, Kawasaki Medical School General Medical Center
| | - Ichiro Morita
- Department of Surgery, Kawasaki Medical School General Medical Center
| | - Atsuhisa Isida
- Department of Surgery, Kawasaki Medical School General Medical Center
| | - Yuka Mikami
- Department of Pathology, Kawasaki Medical School
| | | | - Yasufumi Sato
- New Industry Creation Hatchery Center, Tohoku University
| | | |
Collapse
|
35
|
Hwang J, Kiick KL, Sullivan MO. Modified hyaluronic acid-collagen matrices trigger efficient gene transfer and prohealing behavior in fibroblasts for improved wound repair. Acta Biomater 2022; 150:138-153. [PMID: 35907557 DOI: 10.1016/j.actbio.2022.07.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/01/2022]
Abstract
Growth factor therapy has demonstrated great promise for chronic wound repair, but controlling growth factor activity and cell phenotype over desired time frames remains a critical challenge. In this study, we developed a gene-activated hyaluronic acid-collagen matrix (GAHCM) comprising DNA/polyethylenimine (PEI) polyplexes retained on hyaluronic acid (HA)-collagen hydrogels using collagen mimetic peptides (CMPs). We hypothesized that manipulating both the number of CMP-collagen tethers and the ECM composition would provide a powerful strategy to control growth factor gene transfer kinetics while regulating cell behavior, resulting in enhanced growth factor activity for wound repair. We observed that polyplexes with 50% CMP-modified PEI (50 CP) showed enhanced retention of polyplexes in HCM hydrogels by 2.7-fold as compared to non-CMP modified polyplexes. Moreover, the incorporation of HA in the hydrogel promoted a significant increase in gene transfection efficiency based upon analysis of Gaussia luciferase (GLuc) reporter gene expression, and gene expression could be attenuated by blocking HA-CD44 signaling. Furthermore, when fibroblasts were exposed to vascular endothelial growth factor-A (VEGF-A)-GAHCM, the 50 CP matrix facilitated sustained VEGF-A production for up to 7 days, with maximal expression at day 5. Application of these VEGF-A-50 CP samples stimulated prolonged pro-healing responses, including the TGF-β1-induced myofibroblast-like phenotypes and enhanced closure of murine splinted wounds. Overall, these findings demonstrate the use of ECM-based materials to stimulate efficient gene transfer and regulate cellular phenotype, resulting in improved control of growth factor activity for wound repair. GAHCM have significant potential to overcome key challenges in growth factor therapy for regenerative medicine. STATEMENT OF SIGNIFICANCE: Despite great promise for growth factor therapies in wound treatment, controlling growth factor activity and providing a microenvironment for cells that maximizes growth factor signaling have continued to limit the success of existing formulations. Our GAHCM strategy, combining CMP gene delivery and hyaluronic acid-collagen matrix, enabled enhanced wound healing efficacy via the combination of controlled and localized growth factor expression and matrix-mediated regulation of cell behavior. Incorporation of CMPs and HA in the same matrix synergistically enhanced VEGF activity as compared with simpler matrices. Accordingly, GAHCM will advance our ability to leverage growth factor signaling for wound healing, resulting in new long-term treatments for recalcitrant wounds.
Collapse
Affiliation(s)
- Jeongmin Hwang
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Kristi L Kiick
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA; Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA.
| | - Millicent O Sullivan
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA; Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
36
|
Ciucci G, Colliva A, Vuerich R, Pompilio G, Zacchigna S. Biologics and cardiac disease: challenges and opportunities. Trends Pharmacol Sci 2022; 43:894-905. [PMID: 35779965 DOI: 10.1016/j.tips.2022.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/06/2022] [Accepted: 06/02/2022] [Indexed: 10/31/2022]
Abstract
Biologics are revolutionizing the treatment of chronic diseases, such as cancer and monogenic disorders, by overcoming the limits of classic therapeutic approaches using small molecules. However, the clinical use of biologics is limited for cardiovascular diseases (CVDs) , which are the primary cause of morbidity and mortality worldwide. Here, we review the state-of-the-art use of biologics for cardiac disorders and provide a framework for understanding why they still struggle to enter the field. Some limitations are common and intrinsic to all biological drugs, whereas others depend on the complexity of cardiac disease. In our opinion, delineating these struggles will be valuable in developing and accelerating the approval of a new generation of biologics for CVDs.
Collapse
Affiliation(s)
- Giulio Ciucci
- Cardiovascular Biology Laboratory, ICGEB Trieste, Trieste, Italy
| | - Andrea Colliva
- Cardiovascular Biology Laboratory, ICGEB Trieste, Trieste, Italy; University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Roman Vuerich
- Cardiovascular Biology Laboratory, ICGEB Trieste, Trieste, Italy; University of Trieste, Department of Life Sciences, Trieste, Italy
| | - Giulio Pompilio
- Centro Cardiologico Monzino IRCCS, Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milano, Milano, Italy
| | - Serena Zacchigna
- Cardiovascular Biology Laboratory, ICGEB Trieste, Trieste, Italy; University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy.
| |
Collapse
|
37
|
Grismaldo A, Sobrevia L, Morales L. Role of platelet-derived growth factor c on endothelial dysfunction in cardiovascular diseases. Biochim Biophys Acta Gen Subj 2022; 1866:130188. [PMID: 35691459 DOI: 10.1016/j.bbagen.2022.130188] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/02/2022] [Accepted: 06/05/2022] [Indexed: 01/01/2023]
Abstract
Loss of endothelial function is a common feature to all cardiovascular diseases (CVDs). One of the risk factors associated with the development of CVDs is the hyperglycaemia that occurs in patients with metabolic disorders such as Type 1 and Type 2 diabetes mellitus. Hyperglycaemia causes endothelial dysfunction through increased production of reactive oxygen species (ROS) from different cellular sources leading to oxidative stress. Vascular endothelial growth factor (VEGF) is essential in the stimulation and maintenance of endothelial functional aspects and, although it can mitigate the impact of ROS, VEGF-mediated signalling is partially inhibited in diabetes mellitus. The search for therapeutic strategies that preserve, protect and improve the functions of the endothelium is of great relevance in the investigation of CVDs associated with hyperglycaemia. Platelet-derived growth factor C (PDGF-C) is a peptide with angiogenic properties, independent of VEGF, that stimulates angiogenesis and revascularization of ischemic tissue. In a diabetic mouse model, PDGF-C stimulates mature endothelial cell migration, angiogenesis, endothelial progenitor cell mobilization, and increased neovascularization, and protects blood vessels in a retinal degeneration model activating anti-apoptosis and proliferation signalling pathways in endothelial cells. This review summarizes the information on the damage that high d-glucose causes on endothelial function and the beneficial effects that PDGF-CC could exert in this condition.
Collapse
Affiliation(s)
- Adriana Grismaldo
- Experimental and Computational Biochemistry Group, Faculty of Sciences, Nutrition and Biochemistry Department, Pontificia Universidad Javeriana, Bogotá, DC, Colombia; Cellular and Molecular Physiology Laboratory, Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory, Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; Medical School (Faculty of Medicine), Sao Paulo State University (UNESP), Brazil; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands; Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey, Nuevo León. Mexico..
| | - Ludis Morales
- Experimental and Computational Biochemistry Group, Faculty of Sciences, Nutrition and Biochemistry Department, Pontificia Universidad Javeriana, Bogotá, DC, Colombia.
| |
Collapse
|
38
|
Li X, Wang X, He P, Bennett E, Haggard E, Ma J, Cai C. Mitochondrial Membrane Potential Identifies a Subpopulation of Mesenchymal Progenitor Cells to Promote Angiogenesis and Myocardial Repair. Cells 2022; 11:1713. [PMID: 35626749 PMCID: PMC9139404 DOI: 10.3390/cells11101713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/10/2022] Open
Abstract
Identifying effective donor cells is one of obstacles that limits cell therapy for heart disease. In this study, we sorted a subpopulation of human mesenchymal progenitor cells (hMPCs) from the right atrial appendage using the low mitochondrial membrane potential. Compared to the non-sorted cells, hMPCs hold the capacity for stemness and enrich mesenchymal stem cell markers. The hMPCs display better ability for survival, faster proliferation, less production of reactive oxygen species (ROS), and greater release of cytoprotective cytokines. The hMPCs exhibit decreased expression of senescence genes and increased expression of anti-apoptotic and antioxidant genes. Intramyocardial injection of hMPCs into the infarcted heart resulted in increased left ventricular ejection fraction and reduced cardiac remodeling and infarct size in the group of animals receiving hMPCs. Both in vitro and in vivo studies indicated hMPCs have the potential to differentiate into endothelial cells and smooth muscle cells. Immunohistochemistry staining showed that cell therapy with hMPCs enhances cardiac vascular regeneration and cardiac proliferation, and decreases cardiac cell apoptosis, which is associated with the increased secretion of cytoprotective and pro-angiogenic cytokines. Overall, we discovered a subpopulation of human mesenchymal progenitor cells via their low mitochondrial membrane potential, which might provide an alternative donor cell source for cellular therapy for ischemic heart disease.
Collapse
Affiliation(s)
- Xiuchun Li
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (X.L.); (X.W.); (E.H.); (J.M.)
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA;
| | - Xiaoliang Wang
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (X.L.); (X.W.); (E.H.); (J.M.)
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA;
| | - Pan He
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA;
| | - Edward Bennett
- Division of Cardiothoracic Surgery, Albany Medical Center, Albany, NY 12208, USA;
| | - Erin Haggard
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (X.L.); (X.W.); (E.H.); (J.M.)
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (X.L.); (X.W.); (E.H.); (J.M.)
| | - Chuanxi Cai
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (X.L.); (X.W.); (E.H.); (J.M.)
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA;
| |
Collapse
|
39
|
Kim K, Siddiqui Z, Acevedo-Jake AM, Roy A, Choudhury M, Grasman J, Kumar V. Angiogenic Hydrogels to Accelerate Early Wound Healing. Macromol Biosci 2022; 22:e2200067. [PMID: 35579914 DOI: 10.1002/mabi.202200067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/19/2022] [Indexed: 11/09/2022]
Abstract
The metabolic disorder diabetes mellitus affects an increasing proportion of the population, a number projected to double by 2060. Non-life-threatening comorbidities contribute to an interrupted healing process which is first delayed, then prolonged, and associated with increased susceptibility to infection and sustained and unresolved inflammation. This leads to chronic non-healing wounds and eventually potential amputation of extremities. Here we examine the use of a bioactive angiogenic peptide-based hydrogel, SLan, to improve early wound healing in diabetic rats, and compare its performance to clinically utilized biosynthetic peptide-based materials such as Puramatrix. Streptozotocin-treated diabetic rats underwent 8 mm biopsy wounding in their dorsum to remove the epithelium, adipose tissues and muscle layer of the skin, and served as a model for diabetic wound healing. Wounds were treated with either Low (1w%) SLan, High (4w%) SLan, PBS, Puramatrix or K2 (an unfunctionalized non-bioactive control sequentially similar to SLan), covered with Tegaderm and monitored on days 0, 3, 7, 10, 14, 17, 21, 28; animals were sacrificed for histomorphic analyses and immunostaining. An LC/MS method developed to detect SLan in plasma allows pharmacokinetic analysis showing no trafficking of peptides from the wound site into the circulation. Low and High SLan groups show similar final outcomes of wound contraction as control groups (Puramatrix, PBS and K2). SLan-treated rats, however, show marked improvement in healing in earlier time points, including increased deposition of new mature blood vessels. Additionally, rats in the Low SLan treatment groups showed significantly improved wound contraction over other groups and significantly improved healing in early time points. Altogether our results suggest this material can be used to "jumpstart" the diabetic wound healing process. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- KaKyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Amanda M Acevedo-Jake
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Marwa Choudhury
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Vivek Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.,Department of Biology, New Jersey Institute of Technology, Newark, NJ, 07102, USA.,Department of Chemical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.,Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, 07102, USA
| |
Collapse
|
40
|
H 2O 2-responsive VEGF/NGF gene co-delivery nano-system achieves stable vascularization in ischemic hindlimbs. J Nanobiotechnology 2022; 20:145. [PMID: 35305670 PMCID: PMC8934504 DOI: 10.1186/s12951-022-01328-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/24/2022] [Indexed: 12/02/2022] Open
Abstract
Peripheral vascular disease (PVD) is a common clinical manifestation of atherosclerosis. Vascular endothelial growth factor (VEGF) gene therapy is a promising approach for PVD treatment. However, due to single-gene therapy limitations and high H2O2 pathological microenvironment, VEGF gene therapy are not as expectations and its clinical application are limited. Synergistic effects of Nerve factors and vascular factors in angiogenesis have attracted attention in recent years. In this study, VEGF and nerve growth factor (NGF) genes co-delivery nanoparticles (VEGF/NGF-NPs) were prepared by using H2O2 responsive 6s-PLGA-Po-PEG as a carrier. 6s-PLGA-Po-PEG could react with H2O2 specifically due to the internal peroxalate bond. Angiogenic effects of VEGF/NGF-NPs has been evaluated in cells and hindlimb ischemia mice model. Results showed that VEGF/NGF-NPs promoted VEGF and NGF co-expression simultaneously, eliminated excessive H2O2, strengthened reactions between SH-SY5Ys and HUVECs, and finally enhanced migration, tube formation, proliferation and H2O2 damage resistance of HUVECs. VEGF/NGF-NPs also recovered blood perfusion, promoted the expression of VEGF, NGF, eNOS and NO, and enhanced vascular coverage of pericytes. Treatment effects of VEGF/NGF-NPs may related to VEGF/eNOS/NO pathway. Altogether, VEGF/NGF-NPs eliminated excessive H2O2 while achieving gene co-delivery, and promoted stable angiogenesis. It’s a promising way for PVD treatment by using VEGF/NGF-NPs.
Collapse
|
41
|
Abstract
Obesity has reached epidemic proportions and is a major contributor to insulin resistance (IR) and type 2 diabetes (T2D). Importantly, IR and T2D substantially increase the risk of cardiovascular (CV) disease. Although there are successful approaches to maintain glycemic control, there continue to be increased CV morbidity and mortality associated with metabolic disease. Therefore, there is an urgent need to understand the cellular and molecular processes that underlie cardiometabolic changes that occur during obesity so that optimal medical therapies can be designed to attenuate or prevent the sequelae of this disease. The vascular endothelium is in constant contact with the circulating milieu; thus, it is not surprising that obesity-driven elevations in lipids, glucose, and proinflammatory mediators induce endothelial dysfunction, vascular inflammation, and vascular remodeling in all segments of the vasculature. As cardiometabolic disease progresses, so do pathological changes in the entire vascular network, which can feed forward to exacerbate disease progression. Recent cellular and molecular data have implicated the vasculature as an initiating and instigating factor in the development of several cardiometabolic diseases. This Review discusses these findings in the context of atherosclerosis, IR and T2D, and heart failure with preserved ejection fraction. In addition, novel strategies to therapeutically target the vasculature to lessen cardiometabolic disease burden are introduced.
Collapse
|
42
|
Zogg H, Singh R, Ro S. Current Advances in RNA Therapeutics for Human Diseases. Int J Mol Sci 2022; 23:ijms23052736. [PMID: 35269876 PMCID: PMC8911101 DOI: 10.3390/ijms23052736] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 12/11/2022] Open
Abstract
Following the discovery of nucleic acids by Friedrich Miescher in 1868, DNA and RNA were recognized as the genetic code containing the necessary information for proper cell functioning. In the years following these discoveries, vast knowledge of the seemingly endless roles of RNA have become better understood. Additionally, many new types of RNAs were discovered that seemed to have no coding properties (non-coding RNAs), such as microRNAs (miRNAs). The discovery of these new RNAs created a new avenue for treating various human diseases. However, RNA is relatively unstable and is degraded fairly rapidly once administered; this has led to the development of novel delivery mechanisms, such as nanoparticles to increase stability as well as to prevent off-target effects of these molecules. Current advances in RNA-based therapies have substantial promise in treating and preventing many human diseases and disorders through fixing the pathology instead of merely treating the symptomology similarly to traditional therapeutics. Although many RNA therapeutics have made it to clinical trials, only a few have been FDA approved thus far. Additionally, the results of clinical trials for RNA therapeutics have been ambivalent to date, with some studies demonstrating potent efficacy, whereas others have limited effectiveness and/or toxicity. Momentum is building in the clinic for RNA therapeutics; future clinical care of human diseases will likely comprise promising RNA therapeutics. This review focuses on the current advances of RNA therapeutics and addresses current challenges with their development.
Collapse
|
43
|
Garbern JC, Lee RT. Heart regeneration: 20 years of progress and renewed optimism. Dev Cell 2022; 57:424-439. [PMID: 35231426 PMCID: PMC8896288 DOI: 10.1016/j.devcel.2022.01.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is a leading cause of death worldwide, and thus there remains great interest in regenerative approaches to treat heart failure. In the past 20 years, the field of heart regeneration has entered a renaissance period with remarkable progress in the understanding of endogenous heart regeneration, stem cell differentiation for exogenous cell therapy, and cell-delivery methods. In this review, we highlight how this new understanding can lead to viable strategies for human therapy. For the near term, drugs, electrical and mechanical devices, and heart transplantation will remain mainstays of cardiac therapies, but eventually regenerative therapies based on fundamental regenerative biology may offer more permanent solutions for patients with heart failure.
Collapse
Affiliation(s)
- Jessica C. Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, USA,Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, USA,Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA,Corresponding author and lead contact: Richard T. Lee, Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, Phone: 617-496-5394, Fax: 617-496-8351,
| |
Collapse
|
44
|
Fibrin-based factor delivery for therapeutic angiogenesis: friend or foe? Cell Tissue Res 2022; 387:451-460. [PMID: 35175429 PMCID: PMC8975770 DOI: 10.1007/s00441-022-03598-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/07/2022] [Indexed: 12/28/2022]
Abstract
Therapeutic angiogenesis aims at promoting the growth of blood vessels to restore perfusion in ischemic tissues or aid tissue regeneration. Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis in development, repair, and disease. However, exploiting VEGF for therapeutic purposes has been challenging and needs to take into account some key aspects of VEGF biology. In particular, the spatial localization of angiogenic signals within the extracellular matrix is crucial for physiological assembly and function of new blood vessels. Fibrin is the provisional matrix that is universally deposited immediately after injury and supports the initial steps of tissue regeneration. It provides therefore several ideal features as a substrate to promote therapeutic vascularization, especially through its ability to present growth factors in their physiological matrix-bound state and to modulate their availability for signaling. Here, we provide an overview of fibrin uses as a tissue-engineering scaffold material and as a tunable platform to finely control dose and duration of delivery of recombinant factors in therapeutic angiogenesis. However, in some cases, fibrin has also been associated with undesirable outcomes, namely the promotion of fibrosis and scar formation that actually prevent physiological tissue regeneration. Understanding the mechanisms that tip the balance between the pro- and anti-regenerative functions of fibrin will be the key to fully exploit its therapeutic potential.
Collapse
|
45
|
Vekstein AM, Wendell DC, DeLuca S, Yan R, Chen Y, Bishawi M, Devlin GW, Asokan A, Poss KD, Bowles DE, Williams AR, Bursac N. Targeted Delivery for Cardiac Regeneration: Comparison of Intra-coronary Infusion and Intra-myocardial Injection in Porcine Hearts. Front Cardiovasc Med 2022; 9:833335. [PMID: 35224061 PMCID: PMC8866722 DOI: 10.3389/fcvm.2022.833335] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The optimal delivery route to enhance effectiveness of regenerative therapeutics to the human heart is poorly understood. Direct intra-myocardial (IM) injection is the gold standard, however, it is relatively invasive. We thus compared targeted IM against less invasive, catheter-based intra-coronary (IC) delivery to porcine myocardium for the acute retention of nanoparticles using cardiac magnetic resonance (CMR) imaging and viral vector transduction using qPCR. METHODS Ferumoxytol iron oxide (IO) nanoparticles (5 ml) were administered to Yorkshire swine (n = 13) by: (1) IM via thoracotomy, (2) catheter-based IC balloon-occlusion (BO) with infusion into the distal left anterior descending (LAD) coronary artery, (3) IC perforated side-wall (SW) infusion into the LAD, or (4) non-selective IC via left main (LM) coronary artery infusion. Hearts were harvested and imaged using at 3T whole-body MRI scanner. In separate Yorkshire swine (n = 13), an adeno-associated virus (AAV) vector was similarly delivered, tissue harvested 4-6 weeks later, and viral DNA quantified from predefined areas at risk (apical LV/RV) vs. not at risk in a potential mid-LAD infarct model. Results were analyzed using pairwise Student's t-test. RESULTS IM delivery yielded the highest IO retention (16.0 ± 4.6% of left ventricular volume). Of the IC approaches, BO showed the highest IO retention (8.7 ± 2.2% vs. SW = 5.5 ± 4.9% and LM = 0%) and yielded consistent uptake in the porcine distal LAD territory, including the apical septum, LV, and RV. IM delivery was limited to the apex and anterior wall, without septal retention. For the AAV delivery, the BO was most efficient in the at risk territory (Risk: BO = 6.0 × 10-9, IM = 1.4 × 10-9, LM = 3.2 × 10-10 viral copies per μg genomic DNA) while all delivery routes were comparable in the non-risk territory (BO = 1.7 × 10-9, IM = 8.9 × 10-10, LM = 1.2 × 10-9). CONCLUSIONS Direct IM injection has the highest local retention, while IC delivery with balloon occlusion and distal infusion is the most effective IC delivery technique to target therapeutics to a heart territory most in risk from an infarct.
Collapse
Affiliation(s)
- Andrew M. Vekstein
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - David C. Wendell
- Duke Cardiovascular Magnetic Resonance Center, Duke University Medical Center, Durham, NC, United States
| | - Sophia DeLuca
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
- Department of Cell Biology, Duke Regeneration Center, Duke University, Durham, NC, United States
| | - Ruorong Yan
- Department of Cell Biology, Duke Regeneration Center, Duke University, Durham, NC, United States
| | - Yifan Chen
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Muath Bishawi
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Garth W. Devlin
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, United States
| | - Aravind Asokan
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, United States
| | - Kenneth D. Poss
- Department of Cell Biology, Duke Regeneration Center, Duke University, Durham, NC, United States
| | - Dawn E. Bowles
- Department of Surgery, Surgical Sciences, Duke University Medical Center, Durham, NC, United States
| | - Adam R. Williams
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
46
|
Bupphathong S, Quiroz C, Huang W, Chung PF, Tao HY, Lin CH. Gelatin Methacrylate Hydrogel for Tissue Engineering Applications—A Review on Material Modifications. Pharmaceuticals (Basel) 2022; 15:ph15020171. [PMID: 35215284 PMCID: PMC8878046 DOI: 10.3390/ph15020171] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 11/26/2022] Open
Abstract
To recreate or substitute tissue in vivo is a complicated endeavor that requires biomaterials that can mimic the natural tissue environment. Gelatin methacrylate (GelMA) is created through covalent bonding of naturally derived polymer gelatin and methacrylic groups. Due to its biocompatibility, GelMA receives a lot of attention in the tissue engineering research field. Additionally, GelMA has versatile physical properties that allow a broad range of modifications to enhance the interaction between the material and the cells. In this review, we look at recent modifications of GelMA with naturally derived polymers, nanomaterials, and growth factors, focusing on recent developments for vascular tissue engineering and wound healing applications. Compared to polymers and nanoparticles, the modifications that embed growth factors show better mechanical properties and better cell migration, stimulating vascular development and a structure comparable to the natural-extracellular matrix.
Collapse
Affiliation(s)
- Sasinan Bupphathong
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110, Taiwan; (S.B.); (H.-Y.T.)
| | - Carlos Quiroz
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan;
| | - Wei Huang
- Department of Orthodontics, Rutgers School of Dental Medicine, Newark, NJ 07103, USA;
| | - Pei-Feng Chung
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan;
| | - Hsuan-Ya Tao
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110, Taiwan; (S.B.); (H.-Y.T.)
| | - Chih-Hsin Lin
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110, Taiwan; (S.B.); (H.-Y.T.)
- Correspondence:
| |
Collapse
|
47
|
Costa ALR, Willerth SM, de la Torre LG, Han SW. Trends in hydrogel-based encapsulation technologies for advanced cell therapies applied to limb ischemia. Mater Today Bio 2022; 13:100221. [PMID: 35243296 PMCID: PMC8866736 DOI: 10.1016/j.mtbio.2022.100221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/28/2022] [Accepted: 02/12/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ana Letícia Rodrigues Costa
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas, SP, Brazil
| | - Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8W 2Y2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8W 2Y2, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Lucimara Gaziola de la Torre
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas, SP, Brazil
| | - Sang Won Han
- Department of Biophysics, Escola Paulista de Medicina, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
- Corresponding author.
| |
Collapse
|
48
|
Londhe S, Patra CR. Biomedical applications of europium hydroxide nanorods. Nanomedicine (Lond) 2021; 17:5-8. [PMID: 34873917 DOI: 10.2217/nnm-2021-0351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Swapnali Londhe
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, 500007, Telangana, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, 500007, Telangana, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| |
Collapse
|
49
|
Catar R, Moll G, Kamhieh-Milz J, Luecht C, Chen L, Zhao H, Ernst L, Willy K, Girndt M, Fiedler R, Witowski J, Morawietz H, Ringdén O, Dragun D, Eckardt KU, Schindler R, Zickler D. Expanded Hemodialysis Therapy Ameliorates Uremia-Induced Systemic Microinflammation and Endothelial Dysfunction by Modulating VEGF, TNF-α and AP-1 Signaling. Front Immunol 2021; 12:774052. [PMID: 34858433 PMCID: PMC8632537 DOI: 10.3389/fimmu.2021.774052] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022] Open
Abstract
Abstract Systemic chronic microinflammation and altered cytokine signaling, with adjunct cardiovascular disease (CVD), endothelial maladaptation and dysfunction is common in dialysis patients suffering from end-stage renal disease and associated with increased morbidity and mortality. New hemodialysis filters might offer improvements. We here studied the impact of novel improved molecular cut-off hemodialysis filters on systemic microinflammation, uremia and endothelial dysfunction. Human endothelial cells (ECs) were incubated with uremic serum obtained from patients treated with two different hemodialysis regimens in the Permeability Enhancement to Reduce Chronic Inflammation (PERCI-II) crossover clinical trial, comparing High-Flux (HF) and Medium Cut-Off (MCO) membranes, and then assessed for their vascular endothelial growth factor (VEGF) production and angiogenesis. Compared to HF membranes, dialysis with MCO membranes lead to a reduction in proinflammatory mediators and reduced endothelial VEGF production and angiogenesis. Cytokine multiplex screening identified tumor necrosis factor (TNF) superfamily members as promising targets. The influence of TNF-α and its soluble receptors (sTNF-R1 and sTNF-R2) on endothelial VEGF promoter activation, protein release, and the involved signaling pathways was analyzed, revealing that this detrimental signaling was indeed induced by TNF-α and mediated by AP-1/c-FOS signaling. In conclusion, uremic toxins, in particular TNF-signaling, promote endothelial maladaptation, VEGF expression and aberrant angiogenesis, which can be positively modulated by dialysis with novel MCO membranes. Translational Perspective and Graphical Abstract Systemic microinflammation, altered cytokine signaling, cardiovascular disease, and endothelial maladaptation/dysfunction are common clinical complications in dialysis patients suffering from end-stage renal disease. We studied the impact of novel improved medium-cut-off hemodialysis filters on uremia and endothelial dysfunction. We can show that uremic toxins, especially TNF-signaling, promote endothelial maladaptation, VEGF expression and aberrant angiogenesis, which can be positively modulated by dialysis with novel improved medium-cut-off membranes.
Collapse
Affiliation(s)
- Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Julian Kamhieh-Milz
- Institute of Transfusion Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Christian Luecht
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Lei Chen
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Hongfan Zhao
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Lucas Ernst
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Kevin Willy
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Department of Cardiology, University Hospital Münster, Münster, Germany
| | - Matthias Girndt
- Department of Internal Medicine II, Martin-Luther-University Halle, Halle, Germany
| | - Roman Fiedler
- Department of Internal Medicine II, Martin-Luther-University Halle, Halle, Germany
| | - Janusz Witowski
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Olle Ringdén
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | - Duska Dragun
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Ralf Schindler
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Daniel Zickler
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
50
|
Järveläinen N, Halonen P, Nurro J, Hätinen OP, Korpela H, Mäkinen P, Gan LM, Fritsche-Danielson R, Ylä-Herttuala S. Citrate-Saline-Formulated mRNA Delivery into the Heart Muscle with an Electromechanical Mapping and Injection Catheter Does Not Lead to Therapeutic Effects in a Porcine Chronic Myocardial Ischemia Model. Hum Gene Ther 2021; 32:1295-1307. [PMID: 34494459 DOI: 10.1089/hum.2021.149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Based on recent success in using modified RNA in clinical applications, we tested the safety, feasibility, and efficacy of direct delivery of citrate-saline-formulated mRNA into an hibernating ischemic heart muscle using an electromechanical mapping and injection catheter system (NOGA/Myostar) in a porcine chronic myocardial ischemia model. Chronic ischemia was induced in domestic pigs (n = 24) using a bottleneck stent placed in the left anterior descending coronary artery. Low (1 mg) and high (7.5 mg) doses of citrate-saline-formulated vascular endothelial growth factor (VEGF)-A165 mRNA were administered in the study. LacZ mRNA and citrate-saline buffer were used as controls. Ten intramyocardial injections (200 μL each) of the mRNAs or citrate-saline buffer were given endovascularly into the hibernating ischemic myocardium using the NOGA catheter. Positron emission tomography 15O-radiowater imaging was performed 7 days after the induction of ischemia and 28 days after the mRNA delivery to measure quantitative myocardial blood perfusion. Coronary angiography, left ventricular function measurements, and clinical chemistry were obtained at each time point. Thirty-five days after the mRNA transfers, pigs were sacrificed, and infarct size and general histology were analyzed. LacZ mRNA pigs were sacrificed 24 h after the transduction. Citrate-saline-formulated mRNA delivery into the ischemic myocardium with endovascular injection catheter did not lead to meaningful transduction with the translation of VEGF-A165, nor therapeutic effects in the heart. VEGF-A165 mRNA showed no statistically significant improvements in left ventricular ejection fraction (LVEF), cardiac output, myocardial perfusion, infarct size, collateral growth, or capillary area in the study groups. However, there was a trend in the high-dose group toward an improved LVEF and cardiac output at rest. No significant adverse effects were observed. In conclusion, the NOGA/Myostar injection catheter system is ineffective in delivering citrate-saline-formulated mRNAs into the heart muscle with the doses and methods used in a porcine chronic myocardial ischemia model.
Collapse
Affiliation(s)
- Niko Järveläinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Paavo Halonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jussi Nurro
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Olli-Pekka Hätinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henna Korpela
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Petri Mäkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Li-Ming Gan
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Regina Fritsche-Danielson
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|