1
|
Sun A, Liu S, Yin F, Li Z, Liu Z. Circulating inflammatory cytokines and sarcopenia-related traits: a mendelian randomization analysis. Front Med (Lausanne) 2024; 11:1351376. [PMID: 39193020 PMCID: PMC11347448 DOI: 10.3389/fmed.2024.1351376] [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: 01/22/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Objective To explore the causal relationships between 91 circulating inflammatory cytokines and sarcopenia-related traits (low hand grip strength, appendicular lean mass, and usual walking pace) by Mendelian randomized analysis. Methods Independent genetic variations of inflammatory cytokines and sarcopenia-related traits were selected as instrumental variables from publicly available genome-wide association studies (GWAS). The MR analysis was primarily conducted using the inverse variance-weighted (IVW) method. Sensitivity analyses included Steiger filtering and MR PRESSO, with additional assessments for heterogeneity and pleiotropy. Results The IVW method indicated a causal relationship between Vascular Endothelial Growth Factor A (VEGF-A) and low hand grip strength (OR = 1.05654, 95% CI: 1.02453 to 1.08956, P = 0.00046). Additionally, Tumor Necrosis Factor-beta (TNF-β) was found to have a causal relationship with appendicular lean mass (ALM) (β = 0.04255, 95% CI: 0.02838 to 0.05672, P = 3.96E-09). There was no evidence suggesting a significant causal relationship between inflammatory cytokines and usual walking pace. Conclusion Our research substantiated the causal association between inflammatory cytokines, such as VEGF-A and TNF-β, and sarcopenia. This finding may provide new avenues for future clinical treatments.
Collapse
Affiliation(s)
- Aochuan Sun
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Saiya Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fen Yin
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhuangzhuang Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhengtang Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Weng T, Yang M, Zhang W, Jin R, Xia S, Zhang M, Wu P, He X, Han C, Zhao X, Wang X. Dual gene-activated dermal scaffolds regulate angiogenesis and wound healing by mediating the coexpression of VEGF and angiopoietin-1. Bioeng Transl Med 2023; 8:e10562. [PMID: 37693053 PMCID: PMC10487340 DOI: 10.1002/btm2.10562] [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: 06/30/2022] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 09/12/2023] Open
Abstract
The vascularization of dermal substitutes is a key challenge in efforts to heal deep skin defects. In this study, dual gene-activated dermal scaffolds (DGADSs-1) were fabricated by loading nanocomposite particles of polyethylenimine (PEI)/multiple plasmid DNAs (pDNAs) encoding vascular endothelial growth factor and angiopoietin-1 at a ratio of 1:1. In a similar manner, DGADSs-2 were loaded with a chimeric plasmid encoding both VEGF and Ang-1. In vitro studies showed that both types of DGADSs released PEI/pDNA nanoparticles in a sustained manner; they demonstrated effective transfection ability, leading to upregulated expression of VEGF and Ang-1. Furthermore, both types of DGADSs promoted fibroblast proliferation and blood vessel formation, although DGADSs-1 showed a more obvious promotion effect. A rat full-thickness skin defect model showed that split-thickness skin transplanted using a one-step method could achieve full survival at the 12th day after surgery in both DGADSs-1 and DGADSs-2 groups, and the vascularization time of dermal substitutes was significantly shortened. Compared with the other three groups of scaffolds, the DGADSs-1 group had significantly greater cell infiltration, collagen deposition, neovascularization, and vascular maturation, all of which promoted wound healing. Thus, compared with single-gene-activated dermal scaffolds, DGADSs show greater potential for enhancing angiogenesis. DGADSs with different loading modes also exhibited differences in terms of angiogenesis; the effect of loading two genes (DGADSs-1) was better than the effect of loading a chimeric gene (DGADSs-2). In summary, DGADSs, which continuously upregulate VEGF and Ang-1 expression, offer a new functional tissue-engineered dermal substitute with the ability to activate vascularization.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Min Yang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Wei Zhang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Ronghua Jin
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Sizhan Xia
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Manjia Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Pan Wu
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiaojie He
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Chunmao Han
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiong Zhao
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Xingang Wang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
3
|
VEGF-A and FGF4 Engineered C2C12 Myoblasts and Angiogenesis in the Chick Chorioallantoic Membrane. Biomedicines 2022; 10:biomedicines10081781. [PMID: 35892681 PMCID: PMC9330725 DOI: 10.3390/biomedicines10081781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/06/2022] [Accepted: 07/21/2022] [Indexed: 01/04/2023] Open
Abstract
Angiogenesis is the formation of new blood vessels from pre-existing vessels. Adequate oxygen transport and waste removal are necessary for tissue homeostasis. Restrictions in blood supply can lead to ischaemia which can contribute to disease pathology. Vascular endothelial growth factor (VEGF) is essential in angiogenesis and myogenesis, making it an ideal candidate for angiogenic and myogenic stimulation in muscle. We established C2C12 mouse myoblast cell lines which stably express elevated levels of (i) human VEGF-A and (ii) dual human FGF4-VEGF-A. Both stably transfected cells secreted increased amounts of human VEGF-A compared to non-transfected cells, with the latter greater than the former. In vitro, conditioned media from engineered cells resulted in a significant increase in endothelial cell proliferation, migration, and tube formation. In vivo, this conditioned media produced a 1.5-fold increase in angiogenesis in the chick chorioallantoic membrane (CAM) assay. Delivery of the engineered myoblasts on Matrigel demonstrated continued biological activity by eliciting an almost 2-fold increase in angiogenic response when applied directly to the CAM assay. These studies qualify the use of genetically modified myoblasts in therapeutic angiogenesis for the treatment of muscle diseases associated with vascular defects.
Collapse
|
4
|
Kong L, Li J, Yang Y, Tang H, Zou H. Paeoniflorin alleviates the progression of retinal vein occlusion via inhibiting hypoxia inducible factor-1α/vascular endothelial growth factor/STAT3 pathway. Bioengineered 2022; 13:13622-13631. [PMID: 35653799 PMCID: PMC9275925 DOI: 10.1080/21655979.2022.2081755] [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: 12/01/2022] Open
Abstract
Retinal vein occlusion (RVO) is a severe retinal vascular disease involving several complications, leading to weakening of vision and even blindness. Globally, over 16 million patients with RVO were found in the middle-aged population. Paeoniflorin (PF), a monomer of Taohong Siwu decoction, was reported to exhibit many pharmacological activities including anti-inflammatory, antioxidant, cardioprotective, and neuroprotective effects. However, the effect of PF on the progression of RVO remains unclear. In the current study, CCK8 assay was performed to investigate the cell viability. In addition, transwell assay and western blot were used to measure cell invasion and protein expression, respectively. Moreover, a mouse model of oxygen-induced dischemic retinopathy (OIR) was established. We found PF was able to inhibit the migration and angiogenesis of human retinal capillary endothelial cells under normoxia. Additionally, PF notably prevented hypoxia-induced angiogenesis of human retinal capillary endothelial cells via inhibiting hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF)/STAT3 pathway. Eventually, PF significantly alleviated the retinal lesions in the mouse with OIR. All in all, PF was able to alleviate the progression of retinal vein occlusion via inhibiting HIF-1α/VEGF/STAT3 pathway. These findings might provide some theoretical knowledge for exploring novel effective treatment for patients with RVO.
Collapse
Affiliation(s)
- Lingchun Kong
- Department of Ophthalmology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingjing Li
- Department of Ophthalmology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqin Yang
- Department of Ophthalmology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huixin Tang
- Department of Ophthalmology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong Zou
- Department of Ophthalmology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
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: 1.5] [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
|
6
|
Gatina DZ, Garanina EE, Zhuravleva MN, Synbulatova GE, Mullakhmetova AF, Solovyeva VV, Kiyasov AP, Rutland CS, Rizvanov AA, Salafutdinov II. Proangiogenic Effect of 2A-Peptide Based Multicistronic Recombinant Constructs Encoding VEGF and FGF2 Growth Factors. Int J Mol Sci 2021; 22:ijms22115922. [PMID: 34072943 PMCID: PMC8198600 DOI: 10.3390/ijms22115922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022] Open
Abstract
Coronary artery disease remains one of the primary healthcare problems due to the high cost of treatment, increased number of patients, poor clinical outcomes, and lack of effective therapy. Though pharmacological and surgical treatments positively affect symptoms and arrest the disease progression, they generally exhibit a limited effect on the disease outcome. The development of alternative therapeutic approaches towards ischemic disease treatment, especially of decompensated forms, is therefore relevant. Therapeutic angiogenesis, stimulated by various cytokines, chemokines, and growth factors, provides the possibility of restoring functional blood flow in ischemic tissues, thereby ensuring the regeneration of the damaged area. In the current study, based on the clinically approved plasmid vector pVax1, multigenic constructs were developed encoding vascular endothelial growth factor (VEGF), fibroblast growth factors (FGF2), and the DsRed fluorescent protein, integrated via picornaviruses' furin-2A peptide sequences. In vitro experiments demonstrated that genetically modified cells with engineered plasmid constructs expressed the target proteins. Overexpression of VEGF and FGF2 resulted in increased levels of the recombinant proteins. Concomitantly, these did not lead to a significant shift in the general secretory profile of modified HEK293T cells. Simultaneously, the secretome of genetically modified cells showed significant stimulating effects on the formation of capillary-like structures by HUVEC (endothelial cells) in vitro. Our results revealed that when the multicistronic multigene vectors encoding 2A peptide sequences are created, transient transgene co-expression is ensured. The results obtained indicated the mutual synergistic effects of the growth factors VEGF and FGF2 on the proliferation of endothelial cells in vitro. Thus, recombinant multicistronic multigenic constructs might serve as a promising approach for establishing safe and effective systems to treat ischemic diseases.
Collapse
Affiliation(s)
- Dilara Z. Gatina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.Z.G.); (E.E.G.); (M.N.Z.); (G.E.S.); (A.F.M.); (V.V.S.); (A.P.K.)
| | - Ekaterina E. Garanina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.Z.G.); (E.E.G.); (M.N.Z.); (G.E.S.); (A.F.M.); (V.V.S.); (A.P.K.)
| | - Margarita N. Zhuravleva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.Z.G.); (E.E.G.); (M.N.Z.); (G.E.S.); (A.F.M.); (V.V.S.); (A.P.K.)
| | - Gulnaz E. Synbulatova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.Z.G.); (E.E.G.); (M.N.Z.); (G.E.S.); (A.F.M.); (V.V.S.); (A.P.K.)
| | - Adelya F. Mullakhmetova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.Z.G.); (E.E.G.); (M.N.Z.); (G.E.S.); (A.F.M.); (V.V.S.); (A.P.K.)
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.Z.G.); (E.E.G.); (M.N.Z.); (G.E.S.); (A.F.M.); (V.V.S.); (A.P.K.)
| | - Andrey P. Kiyasov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.Z.G.); (E.E.G.); (M.N.Z.); (G.E.S.); (A.F.M.); (V.V.S.); (A.P.K.)
| | - Catrin S. Rutland
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham LE12 5RD, UK;
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.Z.G.); (E.E.G.); (M.N.Z.); (G.E.S.); (A.F.M.); (V.V.S.); (A.P.K.)
- Correspondence: (A.A.R.); (I.I.S.)
| | - Ilnur I. Salafutdinov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.Z.G.); (E.E.G.); (M.N.Z.); (G.E.S.); (A.F.M.); (V.V.S.); (A.P.K.)
- Correspondence: (A.A.R.); (I.I.S.)
| |
Collapse
|
7
|
Pham‐Nguyen O, Son YJ, Kwon T, Kim J, Jung YC, Park JB, Kang B, Yoo HS. Preparation of Stretchable Nanofibrous Sheets with Sacrificial Coaxial Electrospinning for Treatment of Traumatic Muscle Injury. Adv Healthc Mater 2021; 10:e2002228. [PMID: 33506655 DOI: 10.1002/adhm.202002228] [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: 12/18/2020] [Indexed: 11/09/2022]
Abstract
Traumatic muscle injury with massive loss of muscle volume requires intramuscular implantation of proper scaffolds for fast and successful recovery. Although many artificial scaffolds effectively accelerate formation and maturation of myotubes, limited studies are showing the therapeutic effect of artificial scaffolds in animal models with massive muscle injury. In this study, improved myotube differentiation is approved on stepwise stretched gelatin nanofibers and applied to damaged muscle recovery in an animal model. The gelatin nanofibers are fabricated by a two-step process composed of co-axial electrospinning of poly(ɛ-caprolactone) and gelatin and subsequent removal of the outer shells. When stepwise stretching is applied to the myoblasts on gelatin nanofibers for five days, enhanced myotube formation and polarized elongation are observed. Animal models with volumetric loss at quadriceps femoris muscles (>50%) are transplanted with the myotubes cultivated on thin and flexible gelatin nanofiber. Treated animals more efficiently recover exercising functions of the leg when myotubes and the gelatin nanofiber are co-implanted at the injury sites. This result suggests that mechanically stimulated myotubes on gelatin nanofiber is therapeutically feasible for the robust recovery of volumetric muscle loss.
Collapse
Affiliation(s)
- Oanh‐Vu Pham‐Nguyen
- Department of Biomedical Science Institute of Bioscience and Biotechnology Institute of Molecular Science and Fusion Technology Kangwon National University Chuncheon 24341 Republic of Korea
| | - Young Ju Son
- Department of Biomedical Science Institute of Bioscience and Biotechnology Institute of Molecular Science and Fusion Technology Kangwon National University Chuncheon 24341 Republic of Korea
| | - Tae‐wan Kwon
- Department of Veterinary Surgery, College of Veterinary Medicine and Institute of Veterinary Science Kangwon National University Chuncheon 24341 Republic of Korea
| | - Junhyung Kim
- Department of Veterinary Surgery, College of Veterinary Medicine and Institute of Veterinary Science Kangwon National University Chuncheon 24341 Republic of Korea
| | - Yun Chan Jung
- Chaon 331 Pangyo‐ro Bundang‐gu Seongnam Gyeonggi‐do 13488 Republic of Korea
| | - Jong Bae Park
- Jeonju Center Korea Basic Science Institute Jeonju 54907 Republic of Korea
| | - Byung‐Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine Research Institute for Veterinary Science BK21 PLUS Program for Creative Veterinary Science Research Seoul National University Seoul 08826 Republic of Korea
| | - Hyuk Sang Yoo
- Department of Biomedical Science Institute of Bioscience and Biotechnology Institute of Molecular Science and Fusion Technology Kangwon National University Chuncheon 24341 Republic of Korea
| |
Collapse
|
8
|
Gianni-Barrera R, Di Maggio N, Melly L, Burger MG, Mujagic E, Gürke L, Schaefer DJ, Banfi A. Therapeutic vascularization in regenerative medicine. Stem Cells Transl Med 2020; 9:433-444. [PMID: 31922362 PMCID: PMC7103618 DOI: 10.1002/sctm.19-0319] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Therapeutic angiogenesis, that is, the generation of new vessels by delivery of specific factors, is required both for rapid vascularization of tissue‐engineered constructs and to treat ischemic conditions. Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis. However, uncontrolled expression can lead to aberrant vascular growth and vascular tumors (angiomas). Major challenges to fully exploit VEGF potency for therapy include the need to precisely control in vivo distribution of growth factor dose and duration of expression. In fact, the therapeutic window of VEGF delivery depends on its amount in the microenvironment around each producing cell rather than on the total dose, since VEGF remains tightly bound to extracellular matrix (ECM). On the other hand, short‐term expression of less than about 4 weeks leads to unstable vessels, which promptly regress following cessation of the angiogenic stimulus. Here, we will briefly overview some key aspects of the biology of VEGF and angiogenesis and discuss their therapeutic implications with a particular focus on approaches using gene therapy, genetically modified progenitors, and ECM engineering with recombinant factors. Lastly, we will present recent insights into the mechanisms that regulate vessel stabilization and the switch between normal and aberrant vascular growth after VEGF delivery, to identify novel molecular targets that may improve both safety and efficacy of therapeutic angiogenesis.
Collapse
Affiliation(s)
- Roberto Gianni-Barrera
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Nunzia Di Maggio
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Ludovic Melly
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Cardiac, Vascular, and Thoracic Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Maximilian G Burger
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Plastic and Reconstructive Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Edin Mujagic
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Vascular Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Lorenz Gürke
- Vascular Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Plastic and Reconstructive Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Andrea Banfi
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Plastic and Reconstructive Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland.,Vascular Surgery, Department of Surgery, Basel University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
9
|
Tee JK, Yip LX, Tan ES, Santitewagun S, Prasath A, Ke PC, Ho HK, Leong DT. Nanoparticles' interactions with vasculature in diseases. Chem Soc Rev 2019; 48:5381-5407. [PMID: 31495856 DOI: 10.1039/c9cs00309f] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ever-growing use of inorganic nanoparticles (NPs) in biomedicine provides an exciting approach to develop novel imaging and drug delivery systems, owing to the ease with which these NPs can be functionalized to cater to various applications. In cancer therapeutics, nanomedicine generally relies on the enhanced permeability and retention (EPR) effect observed in tumour vasculature to deliver anti-cancer drugs across the endothelium. However, such a phenomenon is dependent on the tumour microenvironment and is not consistently observed in all tumour types, thereby limiting drug transport to the tumour site. On the other hand, there is a rise in utilizing inorganic NPs to intentionally induce endothelial leakiness, creating a window of opportunity to control drug delivery across the endothelium. While this active targeting approach creates a similar phenomenon compared to the EPR effect arising from tumour tissues, its drug delivery applications extend beyond cancer therapeutics and into other vascular-related diseases. In this review, we summarize the current findings of the EPR effect and assess its limitations in the context of anti-cancer drug delivery systems. While the EPR effect offers a possible route for drug passage, we further explore alternative uses of NPs to create controllable endothelial leakiness within short exposures, a phenomenon we coined as nanomaterial-induced endothelial leakiness (NanoEL). Furthermore, we discuss the main mechanistic features of the NanoEL effect that make it unique from conventionally established endothelial leakiness in homeostatic and pathologic conditions, as well as examine its potential applicability in vascular-related diseases, particularly cancer. Therefore, this new paradigm changes the way inorganic NPs are currently being used for biomedical applications.
Collapse
Affiliation(s)
- Jie Kai Tee
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Biscetti F, Bonadia N, Nardella E, Cecchini AL, Landolfi R, Flex A. The Role of the Stem Cells Therapy in the Peripheral Artery Disease. Int J Mol Sci 2019; 20:E2233. [PMID: 31067647 PMCID: PMC6539394 DOI: 10.3390/ijms20092233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/26/2019] [Accepted: 05/05/2019] [Indexed: 01/08/2023] Open
Abstract
Vascular complications of diabetes mellitus are an important issue for all clinicians involved in the management of this complex pathology. Although many therapeutic advances have been reached, peripheral arterial disease is still an unsolved problem that each year compromises the quality of life and life span of affected patients. Oftentimes, patients, after ineffective attempts of revascularization, undergo greater amputations. At the moment, there is no effective and definitive treatment available. In this scenario, the therapeutic use of stem cells could be an interesting option. The aim of the present review is to gather all the best available evidence in this regard and to define a new role of the stem cells therapy in this field, from biomarker to possible therapeutic target.
Collapse
Affiliation(s)
- Federico Biscetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, U.O.C. Clinica Medica e Malattie Vascolari, 00168 Roma, Italy; (E.N.); andrealeonardo-@hotmail.it (A.L.C.); (R.L.); (A.F.)
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Nicola Bonadia
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
- Fondazione Policlinico Universitario A. Gemelli IRCCS, U.O.C. Medicina d’Urgenza e Pronto Soccorso, 00168 Roma, Italy
| | - Elisabetta Nardella
- Fondazione Policlinico Universitario A. Gemelli IRCCS, U.O.C. Clinica Medica e Malattie Vascolari, 00168 Roma, Italy; (E.N.); andrealeonardo-@hotmail.it (A.L.C.); (R.L.); (A.F.)
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Andrea Leonardo Cecchini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, U.O.C. Clinica Medica e Malattie Vascolari, 00168 Roma, Italy; (E.N.); andrealeonardo-@hotmail.it (A.L.C.); (R.L.); (A.F.)
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Raffaele Landolfi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, U.O.C. Clinica Medica e Malattie Vascolari, 00168 Roma, Italy; (E.N.); andrealeonardo-@hotmail.it (A.L.C.); (R.L.); (A.F.)
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Andrea Flex
- Fondazione Policlinico Universitario A. Gemelli IRCCS, U.O.C. Clinica Medica e Malattie Vascolari, 00168 Roma, Italy; (E.N.); andrealeonardo-@hotmail.it (A.L.C.); (R.L.); (A.F.)
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
11
|
Podkalicka P, Mucha O, Dulak J, Loboda A. Targeting angiogenesis in Duchenne muscular dystrophy. Cell Mol Life Sci 2019; 76:1507-1528. [PMID: 30770952 PMCID: PMC6439152 DOI: 10.1007/s00018-019-03006-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 12/28/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) represents one of the most devastating types of muscular dystrophies which affect boys already at early childhood. Despite the fact that the primary cause of the disease, namely the lack of functional dystrophin is known already for more than 30 years, DMD still remains an incurable disease. Thus, an enormous effort has been made during recent years to reveal novel mechanisms that could provide therapeutic targets for DMD, especially because glucocorticoids treatment acts mostly symptomatic and exerts many side effects, whereas the effectiveness of genetic approaches aiming at the restoration of functional dystrophin is under the constant debate. Taking into account that dystrophin expression is not restricted to muscle cells, but is present also in, e.g., endothelial cells, alterations in angiogenesis process have been proposed to have a significant impact on DMD progression. Indeed, already before the discovery of dystrophin, several abnormalities in blood vessels structure and function have been revealed, suggesting that targeting angiogenesis could be beneficial in DMD. In this review, we will summarize current knowledge about the angiogenesis status both in animal models of DMD as well as in DMD patients, focusing on different organs as well as age- and sex-dependent effects. Moreover, we will critically discuss some approaches such as modulation of vascular endothelial growth factor or nitric oxide related pathways, to enhance angiogenesis and attenuate the dystrophic phenotype. Additionally, we will suggest the potential role of other mediators, such as heme oxygenase-1 or statins in those processes.
Collapse
Affiliation(s)
- Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
12
|
Wang X, Lin M, Kang Y. Engineering Porous β-Tricalcium Phosphate (β-TCP) Scaffolds with Multiple Channels to Promote Cell Migration, Proliferation, and Angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2019; 11:9223-9232. [PMID: 30758175 DOI: 10.1021/acsami.8b22041] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Inadequate oxygen and nutrient diffusion in a porous scaffold often resulted in insufficient formation of branched vasculatures, which hindered bone regeneration. In this study, interconnected porous β-tricalcium phosphate (β-TCP) scaffolds with different geometric designs of channels were fabricated and compared to discover the functionality of structure on facilitating nutrient diffusion for angiogenesis. In vitro fluid transportation and degradation of the scaffolds were performed. Cell infiltration, migration, and proliferation of human umbilical vein endothelial cells (HUVECs) on the scaffolds were carried out under both static and dynamic culture conditions. A computational simulation model and a series of immunofluorescent staining were implemented to understand the mechanism of cell behavior in response to different types of scaffolds. Results showed that geometry with multiple channels significantly accelerated the release of Ca2+ and increased the fluid diffusion efficiency. Moreover, multiple channels promoted HUVECs' infiltration and migration in vitro. The ex vivo implantation results showed that the channels promoted cells from the rats' calvarial bone explants to infiltrate into the implanted scaffold. Multiple channels also stimulated HUVECs' proliferation prominently at both static and dynamic culturing conditions. The expression of both cell migration-related protein α5 and angiogenesis-related protein CD31 on multiple-channeled scaffolds was upregulated compared to that on the other two types of scaffolds, implying that multiple channels reinforced cell migration and angiogenesis. All the findings suggested that the geometric design of multiple channels in the porous β-TCP scaffold has promising potential to promote cell infiltration, migration, and further vascularization when implanted in vivo.
Collapse
Affiliation(s)
| | | | - Yunqing Kang
- Integrative Biology PhD Program, Department of Biological Science, College of Science , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| |
Collapse
|
13
|
He Y, Yu X, Chen Z, Li L. Stromal vascular fraction cells plus sustained release VEGF/Ang-1-PLGA microspheres improve fat graft survival in mice. J Cell Physiol 2018; 234:6136-6146. [PMID: 30238985 DOI: 10.1002/jcp.27368] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 08/16/2018] [Indexed: 01/17/2023]
Abstract
Autologous fat transplantation is increasingly applied in plastic and reconstructive surgery. Stromal vascular fraction cells (SVFs) combined with angiogenic factors, such as VEGF (vascular endothelial growth factor A) and Ang-1 (angiogenin-1), can improve angiogenesis, which is a critical factor for graft survival. However, direct transplant with such a mixture is insufficient owing to the short half-life of angiogenic factors. In this study, we evaluated whether a double sustained release system of VEGF/ANG-1-PLGA (poly (lactic-co-glycolic acid)) microspheres plus SVFs can improve angiogenesis and graft survival after autologous fat transplantation. VEGF/ANG-1-PLGA-sustained release microspheres were fabricated by a modified double emulsion-solvent evaporation technique. Human aspirated fat was mixed with SVF suspension plus VEGF/ANG-1 sustained release microspheres (Group C), SVF suspension (Group B) alone, or Dulbecco's modified Eagle's medium as the control (Group A). Eighteen immunocompromised nude mice were injected with these three mixtures subcutaneously at random positions. After 8 weeks, the mean volume of grafts was greater in the SVFs plus VEGF/ANG-1-PLGA group than in the control and SVFs groups (1.08 ± 0.069 ml vs. 0.62 ± 0.036 ml, and 0.83 ± 0.059 ml, respectively). Histological assessments showed that lower fibrosis, but greater microvascular density in the SVFs plus VEGF/ANG-1-PLGA group than in the other groups, though the SVFs group also had an appropriate capillary density and reduced fibrosis. Our findings indicate that SVFs plus VEGF/ANG-1-PLGA-sustained release microspheres can improve angiogenesis and graft survival after autologous fat transplantation.
Collapse
Affiliation(s)
- Yucang He
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaofang Yu
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhuojie Chen
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liqun Li
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Singh RD, Hillestad ML, Livia C, Li M, Alekseev AE, Witt TA, Stalboerger PG, Yamada S, Terzic A, Behfar A. M 3RNA Drives Targeted Gene Delivery in Acute Myocardial Infarction. Tissue Eng Part A 2018; 25:145-158. [PMID: 30047313 DOI: 10.1089/ten.tea.2017.0445] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
IMPACT STATEMENT The M3RNA (microencapsulated modified messenger RNA) platform is an approach to deliver messenger RNA (mRNA) in vivo, achieving a nonintegrating and viral-free approach to gene therapy. This technology was, in this study, tested for its utility in the myocardium, providing a unique avenue for targeted gene delivery into the freshly infarcted myocardial tissue. This study provides the evidentiary basis for the use of M3RNA in the heart through depiction of its performance in cultured cells, healthy rodent myocardium, and acutely injured porcine hearts. By testing the technology in large animal models of infarction, compatibility of M3RNA with current coronary intervention procedures was verified.
Collapse
Affiliation(s)
- Raman Deep Singh
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Matthew L Hillestad
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher Livia
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota.,3 Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Mark Li
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota.,3 Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Alexey E Alekseev
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota.,4 Institute of Theoretical and Experimental Biophysics, Russian Academy of Science, Moscow, Russia
| | - Tyra A Witt
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Paul G Stalboerger
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Satsuki Yamada
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Andre Terzic
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota.,3 Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Atta Behfar
- 1 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,2 VanCleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota.,3 Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
15
|
Melly L, Cerino G, Frobert A, Cook S, Giraud MN, Carrel T, Tevaearai Stahel HT, Eckstein F, Rondelet B, Marsano A, Banfi A. Myocardial infarction stabilization by cell-based expression of controlled Vascular Endothelial Growth Factor levels. J Cell Mol Med 2018; 22:2580-2591. [PMID: 29478261 PMCID: PMC5908097 DOI: 10.1111/jcmm.13511] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/23/2017] [Indexed: 01/24/2023] Open
Abstract
Vascular Endothelial Growth Factor (VEGF) can induce normal or aberrant angiogenesis depending on the amount secreted in the microenvironment around each cell. Towards a possible clinical translation, we developed a Fluorescence Activated Cell Sorting (FACS)-based technique to rapidly purify transduced progenitors that homogeneously express a desired specific VEGF level from heterogeneous primary populations. Here, we sought to induce safe and functional angiogenesis in ischaemic myocardium by cell-based expression of controlled VEGF levels. Human adipose stromal cells (ASC) were transduced with retroviral vectors and FACS purified to generate two populations producing similar total VEGF doses, but with different distributions: one with cells homogeneously producing a specific VEGF level (SPEC), and one with cells heterogeneously producing widespread VEGF levels (ALL), but with an average similar to that of the SPEC population. A total of 70 nude rats underwent myocardial infarction by coronary artery ligation and 2 weeks later VEGF-expressing or control cells, or saline were injected at the infarction border. Four weeks later, ventricular ejection fraction was significantly worsened with all treatments except for SPEC cells. Further, only SPEC cells significantly increased the density of homogeneously normal and mature microvascular networks. This was accompanied by a positive remodelling effect, with significantly reduced fibrosis in the infarcted area. We conclude that controlled homogeneous VEGF delivery by FACS-purified transduced ASC is a promising strategy to achieve safe and functional angiogenesis in myocardial ischaemia.
Collapse
Affiliation(s)
- Ludovic Melly
- Cell and Gene Therapy, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland.,Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland.,Department of Cardiac Vascular and Thoracic Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Giulia Cerino
- Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| | - Aurélien Frobert
- Department of Cardiology, University of Fribourg, Fribourg, Switzerland
| | - Stéphane Cook
- Department of Cardiology, University of Fribourg, Fribourg, Switzerland
| | | | - Thierry Carrel
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Hendrik T Tevaearai Stahel
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Friedrich Eckstein
- Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| | - Benoît Rondelet
- Department of Cardiac Vascular and Thoracic Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Anna Marsano
- Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| | - Andrea Banfi
- Cell and Gene Therapy, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
16
|
Sharma S, Sapkota D, Xue Y, Rajthala S, Yassin MA, Finne-Wistrand A, Mustafa K. Delivery of VEGFA in bone marrow stromal cells seeded in copolymer scaffold enhances angiogenesis, but is inadequate for osteogenesis as compared with the dual delivery of VEGFA and BMP2 in a subcutaneous mouse model. Stem Cell Res Ther 2018; 9:23. [PMID: 29386057 PMCID: PMC5793460 DOI: 10.1186/s13287-018-0778-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/12/2018] [Accepted: 01/16/2018] [Indexed: 12/19/2022] Open
Abstract
Background In bone tissue engineering (BTE), extensive research into vascular endothelial growth factor A (VEGFA)-mediated angiogenesis has yielded inconsistent results. The aim of this study was to investigate the influence on angio- and osteogenesis of adenoviral-mediated delivery of VEGFA alone or in combination with bone morphogenetic protein 2 (BMP2) in bone marrow stromal cells (BMSC) seeded onto a recently developed poly(LLA-co-CL) scaffold. Methods Human BMSC were engineered to express VEGFA alone or in combination with BMP2 and seeded onto poly(LLA-co-CL) scaffolds. Changes in angiogenic and osteogenic gene and protein levels were examined by quantitative reverse-transcription polymerase chain reaction (RT-PCR), PCR array, and alkaline phosphatase assay. An in vivo subcutaneous mouse model was used to investigate the effect on angio- and osteogenesis of VEGFA alone or in combination with BMP2, using microcomputed tomography (μCT), histology, immunohistochemistry, and immunofluorescence. Results Combined delivery of a lower ratio (1:3) of VEGFA and BMP2 (ad-BMP2 + VEGFA) led to upregulation of osteogenic and angiogenic genes in vitro at 3 and 14 days, compared with mono-delivery of VEGFA (ad-VEGFA) and other controls. In vivo, in a subcutaneous mouse model, both ad-VEGFA and ad-BMP2 + VEGFA scaffold explants exhibited increased angiogenesis at 2 weeks. Enhanced angiogenesis was largely related to the recruitment and differentiation of mouse progenitor cells to the endothelial lineage and, to a lesser extent, to endothelial differentiation of the implanted BMSC. μCT and histological analyses revealed enhanced de novo bone formation only in the ad-BMP2 + VEGFA group, corresponding at the molecular level to the upregulation of genes related to osteogenesis, such as ALPL, RUNX2, and SPP1. Conclusions Although BMSC expressing VEGFA alone or in combination with BMP2 significantly induced angiogenesis, VEGFA alone failed to demonstrate osteogenic activity both in vitro and in vivo. These results not only call into question the use of VEGFA alone in bone regeneration, but also highlight the importance in BTE of appropriately formulated combined delivery of VEGFA and BMP2. Electronic supplementary material The online version of this article (10.1186/s13287-018-0778-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sunita Sharma
- Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen, 5020, Bergen, Norway.
| | - Dipak Sapkota
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, 0316, Oslo, Norway
| | - Ying Xue
- Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen, 5020, Bergen, Norway
| | - Saroj Rajthala
- The Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Mohammed A Yassin
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, 10044, Stockholm, Sweden
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, 10044, Stockholm, Sweden
| | - Kamal Mustafa
- Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen, 5020, Bergen, Norway.
| |
Collapse
|
17
|
Dhindsa DS, Khambhati J, Sandesara PB, Eapen DJ, Quyyumi AA. Biomarkers to Predict Cardiovascular Death. Card Electrophysiol Clin 2017; 9:651-664. [PMID: 29173408 DOI: 10.1016/j.ccep.2017.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This article reviews biomarkers that have been shown to identify subjects at increased risk for cardiovascular death within the general population, in those with established coronary artery disease, and in those with heart failure. Use of biomarkers for risk stratification for sudden cardiac death continues to evolve. It seems that a multimarker strategy for risk stratification using simple measures of circulating proteins and usual clinical risk factors, particularly in patients with known coronary artery disease, can be used to identify patients at near-term risk of death. Whether similar strategies in the general population will prove to be cost-effective needs to be investigated.
Collapse
Affiliation(s)
- Devinder S Dhindsa
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Jay Khambhati
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Pratik B Sandesara
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Danny J Eapen
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA.
| |
Collapse
|
18
|
Song X, Zhang Y, Hou Z, Wu H, Lu S, Tang J, Chen X, Cui H, Li Y, Bi Y, Duan W, Li Z, Li C. Adeno-associated virus serotype 9 mediated vascular endothelial growth factor gene overexpression in mdx mice. Exp Ther Med 2017; 15:1825-1830. [PMID: 29434771 PMCID: PMC5776553 DOI: 10.3892/etm.2017.5610] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/13/2017] [Indexed: 11/05/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal neuromuscular disease caused by the absence of dystrophin. Vascular endothelial growth factor (VEGF) is a heparin-binding dimeric glycoprotein and principal angiogenic factor stimulating the migration, proliferation and expression of various genes in endothelial cells. Recently, VEGF was demonstrated to exhibit an antiapoptotic and direct myogenic effect, as well as to enhance muscle force restoration subsequent to traumatic injury. Therefore, the present study attempted to assess the muscle damage of VEGF overexpression in mdx mice. Adeno-associated virus serotype 9 (AAV9)-VEGF was administered intravenously to mdx mice. At 4 weeks after injection, VEGF was observed to be upregulated in the tibialis anterior muscle. In addition, the serum creatine kinase levels were significantly reduced and fatigue was slowed down, whereas the limb grip strength and weight of mice were markedly increased compared with the saline-treated mdx mice. Furthermore, significantly reduced inflammation and necrosis areas were observed in the muscle tissues of mice in the AAV9-VEGF group. These results suggested that AAV9-mediated VEGF gene overexpression was able to improve the muscle damage in mdx mice.
Collapse
Affiliation(s)
- Xueqin Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China.,Institute of Cardiocerebrovascular Disease, Shijiazhuang, Hebei 050000, P.R. China.,Neurological Laboratory of Hebei, Shijiazhuang, Hebei 050000, P.R. China
| | - Ya Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Zhigang Hou
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Hongran Wu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China.,Neurological Laboratory of Hebei, Shijiazhuang, Hebei 050000, P.R. China
| | - Shan Lu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jin Tang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xuexiao Chen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Hongying Cui
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Yuan Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Yue Bi
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China.,Neurological Laboratory of Hebei, Shijiazhuang, Hebei 050000, P.R. China
| | - Weisong Duan
- Neurological Laboratory of Hebei, Shijiazhuang, Hebei 050000, P.R. China
| | - Zhongyao Li
- Neurological Laboratory of Hebei, Shijiazhuang, Hebei 050000, P.R. China
| | - Chunyan Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China.,Institute of Cardiocerebrovascular Disease, Shijiazhuang, Hebei 050000, P.R. China.,Neurological Laboratory of Hebei, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
19
|
Increased levels of circulating CD34+ cells in neovascular age-related macular degeneration: relation with clinical and OCT features. Eur J Ophthalmol 2017; 28:80-86. [PMID: 28777387 DOI: 10.5301/ejo.5001012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE To investigate the levels of circulating CD34+ stem cells in patients with neovascular type age-related macular degeneration (AMD) and its relation with clinical and optical coherence tomography (OCT) findings. METHODS The study consisted of 55 patients: 28 patients (18 male and 10 female) with neovascular type AMD as a study group and 27 patients (12 male and 15 female) scheduled for cataract surgery as a control group. The level of CD34+ stem cells was measured by flow cytometry. Demographic and clinical data were recorded. RESULTS The mean ages of patients in the study and control groups were 71 ± 8 and 68 ± 6 years, respectively. There was no statistically significant difference in terms of age, sex, or systemic disease association between study and control groups. However, smoking status was significantly higher in the study group (67.9% vs 37.0%; p = 0.02). Stem cell levels were significantly higher in the study group (1.5 ± 0.9 vs 0.5 ± 0.3; p<0.001), but there was no relation between stem cell levels and clinical and OCT findings. CONCLUSIONS Increased circulating CD34+ stem cell levels were observed in patients with choroidal neovascular membrane associated with AMD, but no significant relation was found between cell levels and clinical and OCT findings.
Collapse
|
20
|
|
21
|
Synergistic Effects of Vascular Endothelial Growth Factor on Bone Morphogenetic Proteins Induced Bone Formation In Vivo: Influencing Factors and Future Research Directions. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2869572. [PMID: 28070506 PMCID: PMC5187461 DOI: 10.1155/2016/2869572] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 10/16/2016] [Accepted: 10/24/2016] [Indexed: 02/08/2023]
Abstract
Vascular endothelial growth factor (VEGF) and bone morphogenetic proteins (BMPs), as key mediators in angiogenesis and osteogenesis, are used in a combined delivery manner as a novel strategy in bone tissue engineering. VEGF has the potential to enhance BMPs induced bone formation. Both gene delivery and material-based delivery systems were incorporated in previous studies to investigate the synergistic effects of VEGF and BMPs. However, their results were controversial due to variation of methods incorporated in different studies. Factors influencing the synergistic effects of VEGF on BMPs induced bone formation were identified and analyzed in this review to reduce confusion on this issue. The potential mechanisms and directions of future studies were also proposed here. Further investigating mechanisms of the synergistic effects and optimizing these influencing factors will help to generate more effective bone regeneration.
Collapse
|
22
|
Huard J, Lu A, Mu X, Guo P, Li Y. Muscle Injuries and Repair: What's New on the Horizon! Cells Tissues Organs 2016; 202:227-236. [PMID: 27825155 DOI: 10.1159/000443926] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2016] [Indexed: 11/19/2022] Open
Abstract
Although we recognize the many advantages of improved musculoskeletal health, we also note that our ability to sustain this health and to maintain quality of life in an aging population is currently deficient. However, global efforts have produced numerous advances in tissue engineering and regenerative medicine that will collectively serve to fill this deficiency in the near future. The purpose of this review is to highlight our current knowledge, to outline our recent advances, and to discuss the evolving paradigms in skeletal muscle injury and repair.
Collapse
|
23
|
Chhokar V, Tucker AL. Angiogenesis: Basic Mechanisms and Clinical Applications. Semin Cardiothorac Vasc Anesth 2016. [DOI: 10.1177/108925320300700304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The development and maintenance of an adequate vascular supply is critical for the viability of normal and neoplastic tissues. Angiogenesis, the development of new blood vessels from preexisting capillary networks, plays an important role in a number of physiologic and pathologic processes, including reproduction, wound repair, inflammatory diseases, and tumor growth. Angiogenesis involves sequential steps that are triggered in response to angiogenic growth factors released by inflammatory, mesenchymal, or tumor cells that act as ligands for endothelial cell receptor tyrosine kinases. Stimulated endothelial cells detach from neighboring cells and migrate, proliferate, and form tubes. The immature tubes are subsequently invested and stabilized by pericytes or smooth muscle cells. Angiogenesis depends upon complex interactions among various classes of molecules, including adhesion molecules, proteases, structural proteins, cell surface receptors, and growth factors. The therapeutic manipulation of angiogenesis targeted against ischemic and neoplastic diseases has been investigated in preclinical animal models and in clinical trials. Proangiogenic trials that have stimulated vessel growth in ischemic coronary or peripheral tissues through expression, delivery, or stimulated release of growth factors have shown efficacy in animal models and mixed results in human clinical trials. Antiangiogenic trials have used strategies to block the function of molecules critical for new vessel growth or maturation in the treatment of a variety of malignancies, mostly with results less encouraging than those seen in preclinical models. Pro-and antiangiogenic clinical trials demonstrate that strategies for optimal drug delivery, dosing schedules, patient selection, and endpoint measurements need further investigation and refinement before the therapeutic manipulation of angiogenesis will realize its full clinical potential.
Collapse
Affiliation(s)
- Vikram Chhokar
- Department of Internal Medicine, Salem VA Health System, Roanoke, Virginia
| | - Amy L. Tucker
- Department of Internal Medicine, Cardiovascular Division; Cardiovascular Research Center; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
24
|
Groppa E, Brkic S, Bovo E, Reginato S, Sacchi V, Di Maggio N, Muraro MG, Calabrese D, Heberer M, Gianni-Barrera R, Banfi A. VEGF dose regulates vascular stabilization through Semaphorin3A and the Neuropilin-1+ monocyte/TGF-β1 paracrine axis. EMBO Mol Med 2016; 7:1366-84. [PMID: 26323572 PMCID: PMC4604689 DOI: 10.15252/emmm.201405003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
VEGF is widely investigated for therapeutic angiogenesis, but while short-term delivery is desirable for safety, it is insufficient for new vessel persistence, jeopardizing efficacy. Here, we investigated whether and how VEGF dose regulates nascent vessel stabilization, to identify novel therapeutic targets. Monoclonal populations of transduced myoblasts were used to homogeneously express specific VEGF doses in SCID mouse muscles. VEGF was abrogated after 10 and 17 days by Aflibercept treatment. Vascular stabilization was fastest with low VEGF, but delayed or prevented by higher doses, without affecting pericyte coverage. Rather, VEGF dose-dependently inhibited endothelial Semaphorin3A expression, thereby impairing recruitment of Neuropilin-1-expressing monocytes (NEM), TGF-β1 production and endothelial SMAD2/3 activation. TGF-β1 further initiated a feedback loop stimulating endothelial Semaphorin3A expression, thereby amplifying the stabilizing signals. Blocking experiments showed that NEM recruitment required endogenous Semaphorin3A and that TGF-β1 was necessary to start the Semaphorin3A/NEM axis. Conversely, Semaphorin3A treatment promoted NEM recruitment and vessel stabilization despite high VEGF doses or transient adenoviral delivery. Therefore, VEGF inhibits the endothelial Semaphorin3A/NEM/TGF-β1 paracrine axis and Semaphorin3A treatment accelerates stabilization of VEGF-induced angiogenesis.
Collapse
Affiliation(s)
- Elena Groppa
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Sime Brkic
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Emmanuela Bovo
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Silvia Reginato
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Veronica Sacchi
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Nunzia Di Maggio
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Manuele G Muraro
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Diego Calabrese
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Michael Heberer
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Roberto Gianni-Barrera
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Andrea Banfi
- Department of Biomedicine, University of Basel, Basel, Switzerland Department of Surgery, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
25
|
Belair DG, Miller MJ, Wang S, Darjatmoko SR, Binder BYK, Sheibani N, Murphy WL. Differential regulation of angiogenesis using degradable VEGF-binding microspheres. Biomaterials 2016; 93:27-37. [PMID: 27061268 DOI: 10.1016/j.biomaterials.2016.03.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 10/22/2022]
Abstract
Vascular endothelial growth factor (VEGF) spatial and temporal activity must be tightly controlled during angiogenesis to form perfusable vasculature in a healing wound. The native extracellular matrix (ECM) regulates growth factor activity locally via sequestering, and researchers have used ECM-mimicking approaches to regulate the activity of VEGF in cell culture and in vivo. However, the impact of dynamic, affinity-mediated growth factor sequestering has not been explored in detail with biomaterials. Here, we sought to modulate VEGF activity dynamically over time using poly(ethylene glycol) microspheres containing VEGF-binding peptides (VBPs) and exhibiting varying degradation rates. The degradation rate of VBP microspheres conferred a differential ability to up- or down-regulate VEGF activity in culture with primary human endothelial cells. VBP microspheres with fast-degrading crosslinks reduced VEGF activity and signaling, while VBP microspheres with no inherent degradability sequestered and promoted VEGF activity in culture with endothelial cells. VBP microspheres with degradable crosslinks significantly reduced neovascularization in vivo, but neither non-degradable VBP microspheres nor bolus delivery of soluble VBP reduced neovascularization. The covalent incorporation of VBP to degradable microspheres was required to reduce neovascularization in a mouse model of choroidal neovascularization in vivo, which demonstrates a potential clinical application of degradable VBP microspheres to reduce pathological angiogenesis. The results herein highlight the ability to modulate the activity of a sequestered growth factor by changing the crosslinker identity within PEG hydrogel microspheres. The insights gained here may instruct the design and translation of affinity-based growth factor sequestering biomaterials for regenerative medicine applications.
Collapse
Affiliation(s)
- David G Belair
- Department of Biomedical Engineering, University of Wisconsin-Madison, USA
| | - Michael J Miller
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, USA
| | - Shoujian Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, USA
| | | | | | - Nader Sheibani
- Department of Biomedical Engineering, University of Wisconsin-Madison, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, USA; Material Science Program, University of Wisconsin-Madison, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, USA.
| |
Collapse
|
26
|
Wang X, Wu P, Hu X, You C, Guo R, Shi H, Guo S, Zhou H, Chaoheng Y, Zhang Y, Han C. Polyurethane membrane/knitted mesh-reinforced collagen–chitosan bilayer dermal substitute for the repair of full-thickness skin defects via a two-step procedure. J Mech Behav Biomed Mater 2016; 56:120-133. [DOI: 10.1016/j.jmbbm.2015.11.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/22/2015] [Accepted: 11/25/2015] [Indexed: 01/19/2023]
|
27
|
Long-term safety and stability of angiogenesis induced by balanced single-vector co-expression of PDGF-BB and VEGF164 in skeletal muscle. Sci Rep 2016; 6:21546. [PMID: 26882992 PMCID: PMC4756385 DOI: 10.1038/srep21546] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 01/26/2016] [Indexed: 11/19/2022] Open
Abstract
Therapeutic angiogenesis by growth factor delivery is an attractive treatment strategy for ischemic diseases, yet clinical efficacy has been elusive. The angiogenic master regulator VEGF-A can induce aberrant angiogenesis if expressed above a threshold level. Since VEGF remains localized in the matrix around expressing cells, homogeneous dose distribution in target tissues is required, which is challenging. We found that co-expression of the pericyte-recruiting factor PDGF-BB at a fixed ratio with VEGF from a single bicistronic vector ensured normal angiogenesis despite heterogeneous high VEGF levels. Taking advantage of a highly controlled gene delivery platform, based on monoclonal populations of transduced myoblasts, in which every cell stably produces the same amount of each factor, here we rigorously investigated a) the dose-dependent effects, and b) the long-term safety and stability of VEGF and PDGF-BB co-expression in skeletal muscle. PDGF-BB co-expression did not affect the normal angiogenesis by low and medium VEGF doses, but specifically prevented vascular tumors by high VEGF, yielding instead normal and mature capillary networks, accompanied by robust arteriole formation. Induced angiogenesis persisted unchanged up to 4 months, while no tumors appeared. Therefore, PDGF-BB co-expression is an attractive strategy to improve safety and efficacy of therapeutic angiogenesis by VEGF gene delivery.
Collapse
|
28
|
Almubarak S, Nethercott H, Freeberg M, Beaudon C, Jha A, Jackson W, Marcucio R, Miclau T, Healy K, Bahney C. Tissue engineering strategies for promoting vascularized bone regeneration. Bone 2016; 83:197-209. [PMID: 26608518 PMCID: PMC4911893 DOI: 10.1016/j.bone.2015.11.011] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/06/2015] [Accepted: 11/17/2015] [Indexed: 02/07/2023]
Abstract
This review focuses on current tissue engineering strategies for promoting vascularized bone regeneration. We review the role of angiogenic growth factors in promoting vascularized bone regeneration and discuss the different therapeutic strategies for controlled/sustained growth factor delivery. Next, we address the therapeutic uses of stem cells in vascularized bone regeneration. Specifically, this review addresses the concept of co-culture using osteogenic and vasculogenic stem cells, and how adipose derived stem cells compare to bone marrow derived mesenchymal stem cells in the promotion of angiogenesis. We conclude this review with a discussion of a novel approach to bone regeneration through a cartilage intermediate, and discuss why it has the potential to be more effective than traditional bone grafting methods.
Collapse
Affiliation(s)
- Sarah Almubarak
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Hubert Nethercott
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Marie Freeberg
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Caroline Beaudon
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Amit Jha
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Wesley Jackson
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Kevin Healy
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Chelsea Bahney
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States.
| |
Collapse
|
29
|
Hypoxia-specific, VEGF-expressing neural stem cell therapy for safe and effective treatment of neuropathic pain. J Control Release 2016; 226:21-34. [PMID: 26826306 DOI: 10.1016/j.jconrel.2016.01.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 01/05/2016] [Accepted: 01/26/2016] [Indexed: 12/11/2022]
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic cytokine that stimulates the differentiation and function of vascular endothelial cells. VEGF has been implicated in improving nervous system function after injury. However, uncontrolled overexpression of VEGF increases the risk of tumor formation at the site of gene delivery. For this reason, VEGF expression needs to be strictly controlled. The goal of the present study was to understand the effects of hypoxia-induced gene expression system to control VEGF gene expression in neural stem cells (NSCs) on the regeneration of neural tissue after sciatic nerve injury. In this study, we used the erythropoietin (Epo) enhancer-SV40 promoter system (EpoSV-VEGF-NSCs) for hypoxia-specific VEGF expression. We used three types of NSCs: DsRed-NSCs as controls, SV-VEGF-NSCs as uncontrolled VEGF overexpressing NSCs, and EpoSV-VEGF-NSCs. For comparison of VEGF expression at normoxia and hypoxia, we measured the amount of VEGF secreted. VEGF expression decreased at normoxia and increased at hypoxia for EpoSV-VEGF-NSCs; thus, EpoSV-VEGF-NSCs controlled VEGF expression, dependent upon oxygenation condition. To demonstrate the therapeutic effect of EpoSV-VEGF-NSCs, we transplanted each cell line in a neuropathic pain sciatic nerve injury rat model. The transplanted EpoSV-VEGF-NSCs improved sciatic nerve functional index (SFI), mechanical allodynia, and re-myelination similar to the SV-VEGF-NSCs. Additionally, the number of blood vessels increased to a level similar to that of the SV-VEGF-NSCs. However, we did not observe tumor generation in the EpoSV-VEGF-NSC animals that were unlikely to have tumor formation in the SV-VEGF-NSCs. From our results, we determined that EpoSV-VEGF-NSCs safely regulate VEGF gene expression which is dependent upon oxygenation status. In addition, we found that they are therapeutically appropriate for treating sciatic nerve injury.
Collapse
|
30
|
Kanninen KM, Pomeshchik Y, Leinonen H, Malm T, Koistinaho J, Levonen AL. Applications of the Keap1-Nrf2 system for gene and cell therapy. Free Radic Biol Med 2015; 88:350-361. [PMID: 26164630 DOI: 10.1016/j.freeradbiomed.2015.06.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/23/2015] [Accepted: 06/27/2015] [Indexed: 01/15/2023]
Abstract
Oxidative stress has been implicated to play a role in a number of acute and chronic diseases including acute injuries of the central nervous system, neurodegenerative and cardiovascular diseases, and cancer. The redox-activated transcription factor Nrf2 has been shown to protect many different cell types and organs from a variety of toxic insults, whereas in many cancers, unchecked Nrf2 activity increases the expression of cytoprotective genes and, consequently, provides growth advantage to cancerous cells. Herein, we discuss current preclinical gene therapy approaches to either increase or decrease Nrf2 activity with a special reference to neurological diseases and cancer. In addition, we discuss the role of Nrf2 in stem cell therapy for neurological disorders.
Collapse
Affiliation(s)
- Katja M Kanninen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Yuriy Pomeshchik
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Hanna Leinonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Tarja Malm
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Jari Koistinaho
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland.
| | - Anna-Liisa Levonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland.
| |
Collapse
|
31
|
Grasman JM, Zayas MJ, Page RL, Pins GD. Biomimetic scaffolds for regeneration of volumetric muscle loss in skeletal muscle injuries. Acta Biomater 2015. [PMID: 26219862 DOI: 10.1016/j.actbio.2015.07.038] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Skeletal muscle injuries typically result from traumatic incidents such as combat injuries where soft-tissue extremity injuries are present in one of four cases. Further, about 4.5 million reconstructive surgical procedures are performed annually as a result of car accidents, cancer ablation, or cosmetic procedures. These combat- and trauma-induced skeletal muscle injuries are characterized by volumetric muscle loss (VML), which significantly reduces the functionality of the injured muscle. While skeletal muscle has an innate repair mechanism, it is unable to compensate for VML injuries because large amounts of tissue including connective tissue and basement membrane are removed or destroyed. This results in a significant need to develop off-the-shelf biomimetic scaffolds to direct skeletal muscle regeneration. Here, the structure and organization of native skeletal muscle tissue is described in order to reveal clear design parameters that are necessary for scaffolds to mimic in order to successfully regenerate muscular tissue. We review the literature with respect to the materials and methodologies used to develop scaffolds for skeletal muscle tissue regeneration as well as the limitations of these materials. We further discuss the variety of cell sources and different injury models to provide some context for the multiple approaches used to evaluate these scaffold materials. Recent findings are highlighted to address the state of the field and directions are outlined for future strategies, both in scaffold design and in the use of different injury models to evaluate these materials, for regenerating functional skeletal muscle. STATEMENT OF SIGNIFICANCE Volumetric muscle loss (VML) injuries result from traumatic incidents such as those presented from combat missions, where soft-tissue extremity injuries are represented in one of four cases. These injuries remove or destroy large amounts of skeletal muscle including the basement membrane and connective tissue, removing the structural, mechanical, and biochemical cues that usually direct its repair. This results in a significant need to develop off-the-shelf biomimetic scaffolds to direct skeletal muscle regeneration. In this review, we examine current strategies for the development of scaffold materials designed for skeletal muscle regeneration, highlighting advances and limitations associated with these methodologies. Finally, we identify future approaches to enhance skeletal muscle regeneration.
Collapse
|
32
|
Sicari BM, Londono R, Badylak SF. Strategies for skeletal muscle tissue engineering: seed vs. soil. J Mater Chem B 2015; 3:7881-7895. [PMID: 32262901 DOI: 10.1039/c5tb01714a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The most commonly used tissue engineering approach includes the ex vivo combination of site-appropriate cell(s) and scaffold material(s) to create three-dimensional constructs for tissue replacement or reconstruction. These three-dimensional combinations are typically subjected to a period of culture and conditioning (i.e., self-assembly and maturation) to promote the development of ex vivo constructs which closely mimic native target tissue. This cell-based approach is challenged by the host response to the engineered tissue construct following surgical implantation. As an alternative to the cell-based approach, acellular biologic scaffolds attract endogenous cells and remodel into partially functional mimics of native tissue upon implantation. The present review examines cell-types (i.e., seed), scaffold materials (i.e., soil), and challenges associated with functional tissue engineering. Skeletal muscle is used as the target tissue prototype but the discussed principles will largely apply to most body systems.
Collapse
Affiliation(s)
- Brian M Sicari
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Suite 300, 450 Technology Drive, Pittsburgh, PA 15218, USA.
| | | | | |
Collapse
|
33
|
Split for the cure: VEGF, PDGF-BB and intussusception in therapeutic angiogenesis. Biochem Soc Trans 2015; 42:1637-42. [PMID: 25399582 DOI: 10.1042/bst20140234] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Therapeutic angiogenesis is an attractive strategy to treat patients suffering from ischaemic conditions and vascular endothelial growth factor-A (VEGF) is the master regulator of blood vessel growth. However, VEGF can induce either normal or aberrant angiogenesis depending on its dose localized in the microenvironment around each producing cell in vivo and on the balanced stimulation of platelet-derived growth factor-BB (PDGF-BB) signalling, responsible for pericyte recruitment. At the doses required to induce therapeutic benefit, VEGF causes new vascular growth essentially without sprouting, but rather through the alternative process of intussusception, or vascular splitting. In the present article, we briefly review the therapeutic implications of controlling VEGF dose on one hand and pericyte recruitment on the other, as well as the key features of intussusceptive angiogenesis and its regulation.
Collapse
|
34
|
Takayama K, Kawakami Y, Mifune Y, Matsumoto T, Tang Y, Cummins JH, Greco N, Kuroda R, Kurosaka M, Wang B, Fu FH, Huard J. The effect of blocking angiogenesis on anterior cruciate ligament healing following stem cell transplantation. Biomaterials 2015; 60:9-19. [PMID: 25965282 DOI: 10.1016/j.biomaterials.2015.03.036] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 03/23/2015] [Indexed: 12/30/2022]
Abstract
Ruptured human anterior cruciate ligaments (ACL) contain vascular stem cells capable of enhancing the healing of tendon grafts. In the current study we explored the role that neo-angiogenesis plays in ACL healing. ACL-derived CD34+ cells were isolated via Fluorescence Activated Cell Sorting (FACS) from the rupture sites of human ACLs. The cells were then virally transduced to express either vascular endothelial growth factor (VEGF) or soluble FLT-1 (sFLT-1), which is an antagonist of VEGF. We established five groups: CD34+VEGF(100%), where 100% of the cells were transduced with VEGF, CD34+VEGF(25%), where only 25% of the cells were transduced with VEGF, CD34+, CD34+sFLT-1, and a No cells group. The CD34+sFLT1 group had a significant reduction in biomechanical strength compared to the CD34+ group at 4 and 8 weeks; whereas the biomechanical strength of the CD34+VEGF(25%) group was significantly greater than the CD34+ group at week 4; however, no difference was observed by week 8. Immunohistochemical staining demonstrated a significantly lower number of isolectin B4 and hCD31 positive cells, markers associated with angiogenesis, in the CD34+sFLT1 group, and a higher number of isolectin B4 and hCD31 positive cells in the CD34+VEGF(100%) and CD34+VEGF(25%) groups compared to the CD34+ group. Graft maturation was significantly delayed in the CD34+sFLT1 group and accelerated in the CD34+VEGF(25%) group compared to the CD34+ group. In conclusion, blocking VEGF reduced angiogenesis, graft maturation and biomechanical strength following ACL reconstruction. Native expression of VEGF by the CD34+ cells improved tendon graft maturation and biomechanical strength; however, over-expression of VEGF impeded improvements in biomechanical strength.
Collapse
Affiliation(s)
- Koji Takayama
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yohei Kawakami
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yutaka Mifune
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tomoyuki Matsumoto
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Ying Tang
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - James H Cummins
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Nick Greco
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Masahiro Kurosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Bing Wang
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Freddie H Fu
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
35
|
Martino MM, Brkic S, Bovo E, Burger M, Schaefer DJ, Wolff T, Gürke L, Briquez PS, Larsson HM, Gianni-Barrera R, Hubbell JA, Banfi A. Extracellular matrix and growth factor engineering for controlled angiogenesis in regenerative medicine. Front Bioeng Biotechnol 2015; 3:45. [PMID: 25883933 PMCID: PMC4381713 DOI: 10.3389/fbioe.2015.00045] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/19/2015] [Indexed: 01/22/2023] Open
Abstract
Blood vessel growth plays a key role in regenerative medicine, both to restore blood supply to ischemic tissues and to ensure rapid vascularization of clinical-size tissue-engineered grafts. For example, vascular endothelial growth factor (VEGF) is the master regulator of physiological blood vessel growth and is one of the main molecular targets of therapeutic angiogenesis approaches. However, angiogenesis is a complex process and there is a need to develop rational therapeutic strategies based on a firm understanding of basic vascular biology principles, as evidenced by the disappointing results of initial clinical trials of angiogenic factor delivery. In particular, the spatial localization of angiogenic signals in the extracellular matrix (ECM) is crucial to ensure the proper assembly and maturation of new vascular structures. Here, we discuss the therapeutic implications of matrix interactions of angiogenic factors, with a special emphasis on VEGF, as well as provide an overview of current approaches, based on protein and biomaterial engineering that mimic the regulatory functions of ECM to optimize the signaling microenvironment of vascular growth factors.
Collapse
Affiliation(s)
- Mikaël M Martino
- Host Defense, Immunology Frontier Research Center, Osaka University , Osaka , Japan
| | - Sime Brkic
- Cell and Gene Therapy, Department of Biomedicine, Basel University , Basel , Switzerland ; Department of Surgery, Basel University Hospital , Basel , Switzerland
| | - Emmanuela Bovo
- Cell and Gene Therapy, Department of Biomedicine, Basel University , Basel , Switzerland ; Department of Surgery, Basel University Hospital , Basel , Switzerland
| | - Maximilian Burger
- Cell and Gene Therapy, Department of Biomedicine, Basel University , Basel , Switzerland ; Department of Surgery, Basel University Hospital , Basel , Switzerland ; Plastic, Reconstructive, Aesthetic and Hand Surgery, Department of Surgery, Basel University Hospital , Basel , Switzerland
| | - Dirk J Schaefer
- Plastic, Reconstructive, Aesthetic and Hand Surgery, Department of Surgery, Basel University Hospital , Basel , Switzerland
| | - Thomas Wolff
- Cell and Gene Therapy, Department of Biomedicine, Basel University , Basel , Switzerland ; Department of Surgery, Basel University Hospital , Basel , Switzerland ; Vascular Surgery, Department of Surgery, Basel University Hospital , Basel , Switzerland
| | - Lorenz Gürke
- Vascular Surgery, Department of Surgery, Basel University Hospital , Basel , Switzerland
| | - Priscilla S Briquez
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) , Lausanne , Switzerland
| | - Hans M Larsson
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) , Lausanne , Switzerland
| | - Roberto Gianni-Barrera
- Cell and Gene Therapy, Department of Biomedicine, Basel University , Basel , Switzerland ; Department of Surgery, Basel University Hospital , Basel , Switzerland
| | - Jeffrey A Hubbell
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) , Lausanne , Switzerland ; Institute for Molecular Engineering, University of Chicago , Chicago, IL , USA ; Argonne National Laboratory, Materials Science Division , Argonne, IL , USA
| | - Andrea Banfi
- Cell and Gene Therapy, Department of Biomedicine, Basel University , Basel , Switzerland ; Department of Surgery, Basel University Hospital , Basel , Switzerland
| |
Collapse
|
36
|
Chang KH, Wu YR, Chen YC, Chen CM. Plasma inflammatory biomarkers for Huntington's disease patients and mouse model. Brain Behav Immun 2015; 44:121-7. [PMID: 25266150 DOI: 10.1016/j.bbi.2014.09.011] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/19/2014] [Accepted: 09/20/2014] [Indexed: 12/19/2022] Open
Abstract
Huntington's disease (HD), caused by expanded CAG repeats encoding a polyglutamine tract in the huntingtin (HTT) protein, presents with a predominant degeneration of neurons in the striatum and cortex. Lines of evidence have observed neuroinflammation, particularly microglial activation, is involved in the pathogenesis of HD. Given that HTT is also expressed in peripheral inflammatory cells, it is possible that inflammatory changes detected in peripheral plasma may be biologically relevant and parallel the neuroinflammatory process of HD patients. By examining the expression levels of 13 microglia-derived inflammatory markers in the plasma of 5 PreHD carriers, 15 HD patients and 16 healthy controls, we found plasma levels of IL-6, MMP-9, VEGF and TGF-β1 were significantly increased in HD patients when compared with the controls, while plasma level of IL-18 were significantly reduced in HD patients compared with controls. Plasma level of IL-6 was reversely correlated with the UHDRS independence scale and functional capacity. To understand the temporal correlation between these inflammatory markers and HD progression, their levels were further tested in plasma from R6/2 mouse HD model at different ages. In rotarod test, R6/2 HD mice started to manifest HD phenotype at 7.5 weeks of age. Higher plasma VEGF levels of R6/2 mice than those of age-matched wild-type (WT) littermates were noted from 7 (presymptomatic stage) to 13 weeks of age (late symptomatic stage). The plasma IL-6 levels of R6/2 mice were higher than those of the WT littermates from 9 (early symptomatic stage) to 13 weeks of age. R6/2 mice demonstrated higher MMP-9 and TGF-β1 levels than their WT littermates from 11 (middle symptomatic stage) to 13 weeks of age. In contrast, the plasma IL-18 level was lower than those in WT littermates since 11 weeks of age. These altered expressions of inflammatory markers may serve as the potential biomarkers for HD onset and progression. Specific inhibition/activation of these inflammatory markers may be the targets of HD drug development.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Chun Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
37
|
Won YW, Bull DA, Kim SW. Functional polymers of gene delivery for treatment of myocardial infarct. J Control Release 2014; 195:110-9. [PMID: 25076177 DOI: 10.1016/j.jconrel.2014.07.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/18/2014] [Accepted: 07/20/2014] [Indexed: 01/18/2023]
Abstract
Ischemic heart disease is rapidly growing as the common cause of death in the world. It is a disease that occurs as a result of coronary artery stenosis and is caused by the lack of oxygen within cardiac muscles due to an imbalance between oxygen supply and demand. The conventional medical therapy is focused on the use of drug eluting stents, coronary-artery bypass graft surgery and anti-thrombosis. Gene therapy provides great opportunities for treatment of cardiovascular disease. In order for gene therapy to be successful, the development of proper gene delivery systems and hypoxia-regulated gene expression vectors is the most important factors. Several non-viral gene transfer methods have been developed to overcome the safety problems of viral transduction. Some of which include plasmids that regulate gene expression that is controlled by environment specific promoters in the transcriptional or the translational level. This review explores polymeric gene carriers that target the myocardium and hypoxia-inducible vectors, which regulate gene expression in response to hypoxia, and their application in animal myocardial infarction models.
Collapse
Affiliation(s)
- Young-Wook Won
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - David A Bull
- Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Sung Wan Kim
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
38
|
Shimamura M, Nakagami H, Taniyama Y, Morishita R. Gene therapy for peripheral arterial disease. Expert Opin Biol Ther 2014; 14:1175-84. [PMID: 24766232 DOI: 10.1517/14712598.2014.912272] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Gene therapy has emerged as a novel therapy to promote angiogenesis in patients with critical limb ischemia (CLI) caused by peripheral artery disease. Researchers working in this area have focused on pro-angiogenic factors, such as VEGF, fibroblast growth factor (FGF) and hepatocyte growth factor (HGF). Based on the elaborate studies and favorable results of basic research using naked plasmid DNA (pDNA) encoding these growth factors, some clinical Phase I and Phase II trials have been performed. The results of these studies demonstrate the safety of these approaches and their potential for symptomatic improvement in CLI patients. However, the Phase III clinical trials have so far been limited to HGF gene therapy. Because one pitfall of the Phase III trials has been the limited transgene expression achieved using naked pDNA alone, the development of more efficient gene transfer systems, such as ultrasound microbubbles and the needleless injector, as well as the addition of other genes will make these novel therapies more effective and ease the symptoms of CLI. AREAS COVERED This study reviews the previously published basic research and clinical trials that have studied VEGF, FGF and HGF gene therapies for the treatment of CLI. Adjunctive therapies, such as the addition of prostacyclin synthase genes and the development of more efficient gene transfer techniques for pDNA, are also reviewed. EXPERT OPINION To date, clinical studies have demonstrated the safety of gene therapy in limb ischemia but the effectiveness of this treatment has not been determined. Larger clinical studies, as well as the development of more effective gene therapy, are needed to achieve and confirm beneficial effects.
Collapse
Affiliation(s)
- Munehisa Shimamura
- Osaka University, Kanazawa University and Hamamatsu University School of Medicine, United Graduate School of Child Development, Division of Vascular Medicine and Epigenetics, Department of Child Development , Suita , Japan
| | | | | | | |
Collapse
|
39
|
Sicari BM, Dearth CL, Badylak SF. Tissue Engineering and Regenerative Medicine Approaches to Enhance the Functional Response to Skeletal Muscle Injury. Anat Rec (Hoboken) 2013; 297:51-64. [DOI: 10.1002/ar.22794] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/14/2022]
Affiliation(s)
- Brian M. Sicari
- McGowan Institute for Regenerative Medicine; Pittsburgh Pennsylvania
- Cellular and Molecular Pathology Graduate Program; University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| | - Christopher L. Dearth
- McGowan Institute for Regenerative Medicine; Pittsburgh Pennsylvania
- Department of Surgery; University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine; Pittsburgh Pennsylvania
- Department of Surgery; University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| |
Collapse
|
40
|
Vempati P, Popel AS, Mac Gabhann F. Extracellular regulation of VEGF: isoforms, proteolysis, and vascular patterning. Cytokine Growth Factor Rev 2013; 25:1-19. [PMID: 24332926 DOI: 10.1016/j.cytogfr.2013.11.002] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/14/2013] [Accepted: 11/19/2013] [Indexed: 12/15/2022]
Abstract
The regulation of vascular endothelial growth factor A (VEGF) is critical to neovascularization in numerous tissues under physiological and pathological conditions. VEGF has multiple isoforms, created by alternative splicing or proteolytic cleavage, and characterized by different receptor-binding and matrix-binding properties. These isoforms are known to give rise to a spectrum of angiogenesis patterns marked by differences in branching, which has functional implications for tissues. In this review, we detail the extensive extracellular regulation of VEGF and the ability of VEGF to dictate the vascular phenotype. We explore the role of VEGF-releasing proteases and soluble carrier molecules on VEGF activity. While proteases such as MMP9 can 'release' matrix-bound VEGF and promote angiogenesis, for example as a key step in carcinogenesis, proteases can also suppress VEGF's angiogenic effects. We explore what dictates pro- or anti-angiogenic behavior. We also seek to understand the phenomenon of VEGF gradient formation. Strong VEGF gradients are thought to be due to decreased rates of diffusion from reversible matrix binding, however theoretical studies show that this scenario cannot give rise to lasting VEGF gradients in vivo. We propose that gradients are formed through degradation of sequestered VEGF. Finally, we review how different aspects of the VEGF signal, such as its concentration, gradient, matrix-binding, and NRP1-binding can differentially affect angiogenesis. We explore how this allows VEGF to regulate the formation of vascular networks across a spectrum of high to low branching densities, and from normal to pathological angiogenesis. A better understanding of the control of angiogenesis is necessary to improve upon limitations of current angiogenic therapies.
Collapse
Affiliation(s)
- Prakash Vempati
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Feilim Mac Gabhann
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
41
|
Von Willebrand factor, angiodysplasia and angiogenesis. Mediterr J Hematol Infect Dis 2013; 5:e2013060. [PMID: 24106610 PMCID: PMC3787682 DOI: 10.4084/mjhid.2013.060] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 08/20/2013] [Indexed: 12/21/2022] Open
Abstract
The large multimeric glycoprotein Von Willebrand factor (VWF) is best known for its role in haemostasis; however in recent years other functions of VWF have been identified, indicating that this protein is involved in multiple vascular processes. We recently described a new role for VWF in controlling angiogenesis, which may have significant clinical implications for patients with Von Willebrand disease (VWD), a genetic or acquired condition caused by the deficiency or dysfunction of VWF. VWD can be associated with angiodysplasia, a condition of degenerative blood vessels often present in the gastrointestinal tract, linked to dysregulated angiogenesis. Angiodysplasia can cause severe intractable bleeding, often refractory to conventional VWD treatments. In this review we summarise the evidence showing that VWF controls angiogenesis, and review the angiogenic pathways which have been implicated in this process. We discuss the possible mechanisms though which VWF regulates angiopoietin-2 (Ang-2) and integrin αvβ3, leading to signalling through vascular endothelial growth factor receptor-2 (VEGFR2), one of the most potent activators of angiogenesis. We also review the evidence that links VWF with angiodysplasia, and how the newly identified function of VWF in controlling angiogenesis may pave the way for the development of novel therapies for the treatment of angiodysplasia in congenital VWD and in acquired conditions such as Heyde syndrome.
Collapse
|
42
|
Mujagic E, Gianni-Barrera R, Trani M, Patel A, Gürke L, Heberer M, Wolff T, Banfi A. Induction of aberrant vascular growth, but not of normal angiogenesis, by cell-based expression of different doses of human and mouse VEGF is species-dependent. Hum Gene Ther Methods 2013; 24:28-37. [PMID: 23360398 DOI: 10.1089/hgtb.2012.197] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Therapeutic angiogenesis by vascular endothelial growth factor (VEGF) gene delivery is an attractive approach to treat ischemia. VEGF remains localized around each producing cell in vivo, and overexpression of mouse VEGF(164) (mVEGF(164)) induces normal or aberrant angiogenesis, depending strictly on its dose in the microenvironment in vivo. However, the dose-dependent effects of the clinically relevant factor, human VEGF(165) (hVEGF(165)), are unknown. Here we exploited a highly controlled gene delivery platform, based on clonal populations of transduced myoblasts overexpressing specific VEGF levels, to rigorously compare the in vivo dose-dependent effects of hVEGF(165) and mVEGF(164) in skeletal muscle of severe combined immune deficient (SCID) mice. While low levels of both factors efficiently induced similar amounts of normal angiogenesis, only high levels of mVEGF(164) caused widespread angioma-like structures, whereas equivalent or even higher levels of hVEGF(165) induced exclusively normal and mature capillaries. Expression levels were confirmed both in vitro and in vivo by enzyme-linked immunosorbent assay (ELISA) and quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR). However, in vitro experiments showed that hVEGF(165) was significantly more effective in activating VEGF receptor signaling in human endothelial cells than mVEGF(164), while the opposite was true in murine endothelial cells. In conclusion, we found that, even though hVEGF is similarly efficient to the syngenic mVEGF in inducing angiogenesis at lower doses in a widely adopted and convenient mouse preclinical model, species-dependent differences in the relative activation of the respective receptors may specifically mask the toxic effects of high doses of the human factor.
Collapse
Affiliation(s)
- Edin Mujagic
- Cell and Gene Therapy, Department of Biomedicine and Department of Surgery, Basel University Hospital and Basel University, Basel CH-4031, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Wang X, You C, Hu X, Zheng Y, Li Q, Feng Z, Sun H, Gao C, Han C. The roles of knitted mesh-reinforced collagen-chitosan hybrid scaffold in the one-step repair of full-thickness skin defects in rats. Acta Biomater 2013; 9:7822-32. [PMID: 23603532 DOI: 10.1016/j.actbio.2013.04.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 03/21/2013] [Accepted: 04/09/2013] [Indexed: 12/18/2022]
Abstract
Full-thickness skin defects represent a significant and urgent clinical problem. Dermal substitutes serving as a regenerative template to induce dermal reconstruction provide a promising method to treat serious skin defects. Although collagen-chitosan dermal scaffolds display good biocompatibility and a suitable porous structure for angiogenesis and tissue regeneration, their poor mechanical properties compromise their application. To develop a well-supported dermal substitute, a poly(l-lactide-co-glycolide) (PLGA) knitted mesh was fabricated and integrated with collagen-chitosan scaffold (CCS) to obtain a PLGA knitted mesh-reinforced CCS (PLGAm/CCS). The morphology of this PLGAm/CCS was investigated in vitro. To characterize the tissue response, specifically angiogenesis and tissue regeneration, the PLGAm/CCS was transplanted in combination with thin split-thickness autografts to repair full-thickness skin wounds using a one-step surgical procedure in Sprague-Dawley rats. These results were then compared with CCSs. At weeks 2, 4 and 8 after the operation, the healing wounds were imaged to analyse wound changes, and tissue specimens were harvested for histology, immunohistochemistry, real-time quantitative polymerase chain reaction and Western blot analysis. The results demonstrated that collagen-chitosan sponge in the PLGAm/CCS remained porous, interconnected and occupied the openings of PLGA mesh, and the incorporation of the PLGA knitted mesh into CCS improved the mechanical strength with little influence on its mean pore size and porosity. Following transplantation, PLGAm/CCS inhibited wound contraction, and effectively promoted neotissue formation and blood vessel ingrowth. In conclusion, the mechanical strength of the scaffolds plays an important role in the process of tissue regeneration and vascularization. The ability of PLGAm/CCS to promote angiogenesis and induce in situ tissue regeneration demonstrates its potential in skin tissue engineering.
Collapse
Affiliation(s)
- Xingang Wang
- Department of Burns, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou 310009, China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jazwa A, Florczyk U, Jozkowicz A, Dulak J. Gene therapy on demand: Site specific regulation of gene therapy. Gene 2013; 525:229-38. [DOI: 10.1016/j.gene.2013.03.093] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/02/2013] [Accepted: 03/07/2013] [Indexed: 12/29/2022]
|
45
|
Melly LF, Marsano A, Frobert A, Boccardo S, Helmrich U, Heberer M, Eckstein FS, Carrel TP, Giraud MN, Tevaearai HT, Banfi A. Controlled angiogenesis in the heart by cell-based expression of specific vascular endothelial growth factor levels. Hum Gene Ther Methods 2013; 23:346-56. [PMID: 23075102 DOI: 10.1089/hgtb.2012.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) can induce normal angiogenesis or the growth of angioma-like vascular tumors depending on the amount secreted by each producing cell because it remains localized in the microenvironment. In order to control the distribution of VEGF expression levels in vivo, we recently developed a high-throughput fluorescence-activated cell sorting (FACS)-based technique to rapidly purify transduced progenitors that homogeneously express a specific VEGF dose from a heterogeneous primary population. Here we tested the hypothesis that cell-based delivery of a controlled VEGF level could induce normal angiogenesis in the heart, while preventing the development of angiomas. Freshly isolated human adipose tissue-derived stem cells (ASC) were transduced with retroviral vectors expressing either rat VEGF linked to a FACS-quantifiable cell-surface marker (a truncated form of CD8) or CD8 alone as control (CTR). VEGF-expressing cells were FACS-purified to generate populations producing either a specific VEGF level (SPEC) or uncontrolled heterogeneous levels (ALL). Fifteen nude rats underwent intramyocardial injection of 10(7) cells. Histology was performed after 4 weeks. Both the SPEC and ALL cells produced a similar total amount of VEGF, and both cell types induced a 50%-60% increase in both total and perfused vessel density compared to CTR cells, despite very limited stable engraftment. However, homogeneous VEGF expression by SPEC cells induced only normal and stable angiogenesis. Conversely, heterogeneous expression of a similar total amount by the ALL cells caused the growth of numerous angioma-like structures. These results suggest that controlled VEGF delivery by FACS-purified ASC may be a promising strategy to achieve safe therapeutic angiogenesis in the heart.
Collapse
Affiliation(s)
- Ludovic F Melly
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, 4031 Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Shin SH, Lee J, Ahn DG, Lee KY. Co-delivery of Vascular Endothelial Growth Factor and Angiopoietin-1 Using Injectable Microsphere/Hydrogel Hybrid Systems for Therapeutic Angiogenesis. Pharm Res 2013; 30:2157-65. [DOI: 10.1007/s11095-013-1076-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 05/08/2013] [Indexed: 11/30/2022]
|
47
|
Ennen JP, Verma M, Asakura A. Vascular-targeted therapies for Duchenne muscular dystrophy. Skelet Muscle 2013; 3:9. [PMID: 23618411 PMCID: PMC3651321 DOI: 10.1186/2044-5040-3-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/25/2013] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy and an X-linked recessive, progressive muscle wasting disease caused by the absence of a functional dystrophin protein. Dystrophin has a structural role as a cytoskeletal stabilization protein and protects cells against contraction-induced damage. Dystrophin also serves a signaling role through mechanotransduction of forces and localization of neuronal nitric oxide synthase (nNOS), which produces nitric oxide (NO) to facilitate vasorelaxation. In DMD, the signaling defects produce inadequate tissue perfusion caused by functional ischemia due to a diminished ability to respond to shear stress induced endothelium-dependent dilation. Additionally, the structural defects seen in DMD render myocytes with an increased susceptibility to mechanical stress. The combination of both defects is necessary to generate myocyte damage, which induces successive rounds of myofiber degeneration and regeneration, loss of calcium homeostasis, chronic inflammatory response, fibrosis, and myonecrosis. In individuals with DMD, these processes inevitably cause loss of ambulation shortly after the first decade and an abbreviated life with death in the third or fourth decade due to cardio-respiratory anomalies. There is no known cure for DMD, and although the culpable gene has been identified for more than twenty years, research on treatments has produced few clinically relevant results. Several recent studies on novel DMD therapeutics are vascular targeted and focused on attenuating the inherent functional ischemia. One approach improves vasorelaxation capacity through pharmaceutical inhibition of either phosphodiesterase 5 (PDE5) or angiotensin-converting enzyme (ACE). Another approach increases the density of the underlying vascular network by inducing angiogenesis, and this has been accomplished through either direct delivery of vascular endothelial growth factor (VEGF) or by downregulating the VEGF decoy-receptor type 1 (VEGFR-1 or Flt-1). The pro-angiogenic approaches also seem to be pro-myogenic and could resolve the age-related decline in satellite cell (SC) quantity seen in mdx models through expansion of the SC juxtavascular niche. Here we review these four vascular targeted treatment strategies for DMD and discuss mechanisms, proof of concept, and the potential for clinical relevance associated with each therapy.
Collapse
Affiliation(s)
- James P Ennen
- Stem Cell Institute, University of Minnesota Medical School, McGuire Translational Research Facility, Room 4-220, 2001 6th Street SE, Minneapolis, MN 55455, USA.
| | | | | |
Collapse
|
48
|
Fang Q, Mok PY, Thomas AE, Haddad DJ, Saini SA, Clifford BT, Kapasi NK, Danforth OM, Usui M, Ye W, Luu E, Sharma R, Bartel MJ, Pathmanabhan JA, Ang AAS, Sievers RE, Lee RJ, Springer ML. Pleiotrophin gene therapy for peripheral ischemia: evaluation of full-length and truncated gene variants. PLoS One 2013; 8:e61413. [PMID: 23630585 PMCID: PMC3632611 DOI: 10.1371/journal.pone.0061413] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 03/10/2013] [Indexed: 11/19/2022] Open
Abstract
Pleiotrophin (PTN) is a growth factor with both pro-angiogenic and limited pro-tumorigenic activity. We evaluated the potential for PTN to be used for safe angiogenic gene therapy using the full length gene and a truncated gene variant lacking the domain implicated in tumorigenesis. Mouse myoblasts were transduced to express full length or truncated PTN (PTN or T-PTN), along with a LacZ reporter gene, and injected into mouse limb muscle and myocardium. In cultured myoblasts, PTN was expressed and secreted via the Golgi apparatus, but T-PTN was not properly secreted. Nonetheless, no evidence of uncontrolled growth was observed in cells expressing either form of PTN. PTN gene delivery to myocardium, and non-ischemic skeletal muscle, did not result in a detectable change in vascularity or function. In ischemic hindlimb at 14 days post-implantation, intramuscular injection with PTN-expressing myoblasts led to a significant increase in skin perfusion and muscle arteriole density. We conclude that (1) delivery of the full length PTN gene to muscle can be accomplished without tumorigenesis, (2) the truncated PTN gene may be difficult to use in a gene therapy context due to inefficient secretion, (3) PTN gene delivery leads to functional benefit in the mouse acute ischemic hindlimb model.
Collapse
Affiliation(s)
- Qizhi Fang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Pamela Y. Mok
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Anila E. Thomas
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Daniel J. Haddad
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Shereen A. Saini
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Brian T. Clifford
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Neel K. Kapasi
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Olivia M. Danforth
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Minako Usui
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Weisheng Ye
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Emmy Luu
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Rikki Sharma
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Maya J. Bartel
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Jeremy A. Pathmanabhan
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Andrew A. S. Ang
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Richard E. Sievers
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
| | - Randall J. Lee
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California, United States of America
| | - Matthew L. Springer
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
- Division of Cardiology, University of California San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
49
|
Zhou W, He DQ, Liu JY, Feng Y, Zhang XY, Hua CG, Tang XF. Angiogenic gene-modified myoblasts promote vascularization during repair of skeletal muscle defects. J Tissue Eng Regen Med 2013; 9:1404-16. [PMID: 23365046 DOI: 10.1002/term.1692] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 10/20/2012] [Accepted: 12/20/2012] [Indexed: 02/05/2023]
Affiliation(s)
- Wei Zhou
- Department of Head and Neck Oncology; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Deng-Qi He
- Department of Head and Neck Oncology; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Ji-Yuan Liu
- Department of Head and Neck Oncology; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Yang Feng
- Department of Head and Neck Oncology; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Xiang-Yu Zhang
- Department of Head and Neck Oncology; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Cheng-Ge Hua
- Department of Head and Neck Oncology; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Xiu-Fa Tang
- Department of Head and Neck Oncology; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
50
|
Rhim T, Lee DY, Lee M. Drug delivery systems for the treatment of ischemic stroke. Pharm Res 2013; 30:2429-44. [PMID: 23307348 DOI: 10.1007/s11095-012-0959-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 12/07/2012] [Indexed: 12/13/2022]
Abstract
Stroke is the third leading cause of death in the United States. Reduced cerebral blood flow causes acute damage to the brain due to excitotoxicity, reactive oxygen species (ROS), and ischemia. Currently, the main treatment for stroke is to revive the blood flow by using thrombolytic agents. Reviving blood flow also causes ischemia-reperfusion (I/R) damage. I/R damage results from inflammation and apoptosis and can persist for days to weeks, increasing the infarct size. Drugs can be applied to stroke to intervene in the sub-acute and chronic phases. Chemical, peptide, and genetic therapies have been evaluated to reduce delayed damage to the brain. These drugs have different characteristics, requiring that delivery carriers be developed based on these characteristics. The delivery route is another important factor affecting the efficiency of drug delivery. Various delivery routes have been developed, such as intravenous injection, intranasal administration, and local direct injection to overcome the blood-brain-barrier (BBB). In this review, the delivery carriers and delivery routes for peptide and gene therapies are discussed and examples are provided. Combined with new drugs, drug delivery systems will eventually provide useful treatments for ischemic stroke.
Collapse
Affiliation(s)
- Taiyoun Rhim
- Department of Bioengineering, College of Engineering, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | | | | |
Collapse
|