1
|
Basuthakur P, Roy A, Ghosh S, Vijay V, Sinha D, Radhakrishnan M, Kumar A, Patra CR, Chakravarty S. Pro-angiogenic Terbium Hydroxide Nanorods Improve Critical Limb Ischemia in Part by Amelioration of Ischemia-Induced Endothelial Injury. ACS APPLIED BIO MATERIALS 2024; 7:4389-4405. [PMID: 38848346 DOI: 10.1021/acsabm.4c00252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Critical limb ischemia (CLI) refers to a severe condition resulting from gradual obstruction in the supply of blood, oxygen, and nutrients to the limbs. The most promising clinical solution to CLI is therapeutic angiogenesis. This study explored the potency of pro-angiogenic terbium hydroxide nanorods (THNR) for treatment of CLI, with a major focus on their impact on ischemia-induced maladaptive alterations in endothelial cells as well as on vascularization in ischemic limbs. This study demonstrated that, in hypoxia-exposed endothelial cells, THNR improve survival and promote proliferation, migration, restoration of nitric oxide production, and regulation of vascular permeability. Based on molecular studies, these attributes of THNR can be traced to the stimulation of PI3K/AKT/eNOS signaling pathways. Besides, Wnt/GSK-3β/β-catenin signaling pathways may also play a role in the therapeutic actions of THNR. Furthermore, in the murine model of CLI, THNR administration can integrally re-establish blood perfusion with concomitant reduction of muscle damage and inflammation. Additionally, improvement of locomotor activities and motor coordination in ischemic limbs in THNR treated mice is also evident. Overall, the study demonstrates that THNR have the potential to be developed as an efficacious and cost-effective alternative clinical therapy for CLI, using a nanomedicine approach.
Collapse
Affiliation(s)
- Papia Basuthakur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Arpita Roy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Soumya Ghosh
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vincy Vijay
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debiprasad Sinha
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mydhili Radhakrishnan
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Arvind Kumar
- Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumana Chakravarty
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
2
|
Ueno K, Kurazumi H, Suzuki R, Yanagihara M, Mizoguchi T, Harada T, Morikage N, Hamano K. miR-709 exerts an angiogenic effect through a FGF2 upregulation induced by a GSK3B downregulation. Sci Rep 2024; 14:11372. [PMID: 38762650 PMCID: PMC11102560 DOI: 10.1038/s41598-024-62340-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/15/2024] [Indexed: 05/20/2024] Open
Abstract
The aim of this study was to identify angiogenic microRNAs (miRNAs) that could be used in the treatment of hindlimb ischemic tissues. miRNAs contained in extracellular vesicles (EVs) deriving from the plasma were analyzed in C57BL/6 mice, which have ischemia tolerance, and in BALB/c mice without ischemia tolerance as part of a hindlimb ischemia model; as a result 43 angiogenic miRNA candidates were identified. An aortic ring assay was employed by using femoral arteries isolated from BALC/c mice and EVs containing miRNA; as a result, the angiogenic miRNA candidates were limited to 14. The blood flow recovery was assessed after injecting EVs containing miRNA into BALB/c mice with hindlimb ischemia, and miR-709 was identified as a promising angiogenic miRNA. miR-709-encapsulating EVs were found to increase the expression levels of the fibroblast growth factor 2 (FGF2) mRNA in the thigh tissues of hindlimb ischemia model BALB/c mice. miR-709 was also found to bind to the 3'UTR of glycogen synthase kinase 3 beta (GSK3B) in three places. GSK3B-knockdown human artery-derived endothelial cells were found to express high levels of FGF2, and were characterized by increased cell proliferation. These findings indicate that miR-709 induces an upregulation of FGF2 through the downregulation of GSK3B.
Collapse
Affiliation(s)
- Koji Ueno
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan.
- Division of Advanced Cell Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi, Japan.
| | - Hiroshi Kurazumi
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Ryo Suzuki
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Masashi Yanagihara
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Takahiro Mizoguchi
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Takasuke Harada
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Noriyasu Morikage
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
- Division of Advanced Cell Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi, Japan
| |
Collapse
|
3
|
Dhanjal DS, Singh R, Sharma V, Nepovimova E, Adam V, Kuca K, Chopra C. Advances in Genetic Reprogramming: Prospects from Developmental Biology to Regenerative Medicine. Curr Med Chem 2024; 31:1646-1690. [PMID: 37138422 DOI: 10.2174/0929867330666230503144619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 05/05/2023]
Abstract
The foundations of cell reprogramming were laid by Yamanaka and co-workers, who showed that somatic cells can be reprogrammed into pluripotent cells (induced pluripotency). Since this discovery, the field of regenerative medicine has seen advancements. For example, because they can differentiate into multiple cell types, pluripotent stem cells are considered vital components in regenerative medicine aimed at the functional restoration of damaged tissue. Despite years of research, both replacement and restoration of failed organs/ tissues have remained elusive scientific feats. However, with the inception of cell engineering and nuclear reprogramming, useful solutions have been identified to counter the need for compatible and sustainable organs. By combining the science underlying genetic engineering and nuclear reprogramming with regenerative medicine, scientists have engineered cells to make gene and stem cell therapies applicable and effective. These approaches have enabled the targeting of various pathways to reprogramme cells, i.e., make them behave in beneficial ways in a patient-specific manner. Technological advancements have clearly supported the concept and realization of regenerative medicine. Genetic engineering is used for tissue engineering and nuclear reprogramming and has led to advances in regenerative medicine. Targeted therapies and replacement of traumatized , damaged, or aged organs can be realized through genetic engineering. Furthermore, the success of these therapies has been validated through thousands of clinical trials. Scientists are currently evaluating induced tissue-specific stem cells (iTSCs), which may lead to tumour-free applications of pluripotency induction. In this review, we present state-of-the-art genetic engineering that has been used in regenerative medicine. We also focus on ways that genetic engineering and nuclear reprogramming have transformed regenerative medicine and have become unique therapeutic niches.
Collapse
Affiliation(s)
- Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Varun Sharma
- Head of Bioinformatic Division, NMC Genetics India Pvt. Ltd., Gurugram, India
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, CZ 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, Brno, CZ-612 00, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, 50005, Czech Republic
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
4
|
Gene Therapy and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:235-254. [DOI: 10.1007/978-981-19-5642-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
5
|
Salyers ZR, Coleman M, Le D, Ryan TE. AAV-mediated expression of PFKFB3 in myofibers, but not endothelial cells, improves ischemic muscle function in mice with critical limb ischemia. Am J Physiol Heart Circ Physiol 2022; 323:H424-H436. [PMID: 35867710 PMCID: PMC11834898 DOI: 10.1152/ajpheart.00121.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 01/18/2023]
Abstract
6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) is a powerful driver of angiogenesis through its modulation of glycolytic metabolism within endothelial cells. Recent work has demonstrated that PFKFB3 modulates the response to muscle ischemia, however the cell specificity of these effects is not fully understood. In this study, we tested the impact of viral mediated expression of PFKFB3, driven by gene promoters specific for myofibers or endothelial cells, on ischemic hindlimb revascularization and muscle function. We hypothesized that both endothelium- and muscle-specific expression of PFKFB3 would attenuate limb pathology following femoral artery ligation. Male and female BALB/cJ mice were injected with adeno-associated virus encoding the either a green fluorescent protein (GFP) or PFKFB3 driven by either the human skeletal actin (ACTA1) or cadherin-5 (Cdh5) promoters. Four weeks after AAV treatment, mice were subjected to unilateral femoral artery ligation and limb perfusion and muscle function were assessed. Both endothelium- and muscle-specific PFKFB3 expression resulted in significantly more perfused capillaries within the ischemic limb muscle, but neither changed myofiber size/area. Muscle-specific, but not endothelium-specific, PFKFB3 expression significantly improved maximal force production in ischemic muscle (P = 0.0005). Notably, there was a significant effect of sex on maximal force levels in both cohorts of mice (P = 0.0075 and P = 0.0481), indicating that female mice had higher ischemic muscle strength compared with male mice, regardless of treatment group. Taken together, these data demonstrate that although both muscle- and endothelium-specific expression of PFKFB3 enhanced ischemic revascularization, only muscle-specific PFKFB3 expression improved muscle function.NEW & NOTEWORTHY Critical limb ischemia (CLI) carries a significant risk for limb amputation, and treatment options remain limited. We tested the impact of expression of PFKFB3 in myofibers or endothelial cells on limb pathology in mice with CLI. Although both muscle and endothelium-specific PFKFB3 expression increased perfused capillary density, only muscle-specific PFKFB3 expression improve contractile function. Regardless of treatment, female mice demonstrated better recovery from limb ischemic compared with male mice.
Collapse
Affiliation(s)
- Zachary R. Salyers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | - Madeline Coleman
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | - Dennis Le
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
- Center for Exercise Science, University of Florida, Gainesville, FL
- Myology Institute, University of Florida, Gainesville, FL
| |
Collapse
|
6
|
Pan Y, Luo Y, Hong J, He H, Dai L, Zhu H, Wu J. Advances for the treatment of lower extremity arterial disease associated with diabetes mellitus. Front Mol Biosci 2022; 9:929718. [PMID: 36060247 PMCID: PMC9429832 DOI: 10.3389/fmolb.2022.929718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Lower extremity arterial disease (LEAD) is a major vascular complication of diabetes. Vascular endothelial cells dysfunction can exacerbate local ischemia, leading to a significant increase in amputation, disability, and even mortality in patients with diabetes combined with LEAD. Therefore, it is of great clinical importance to explore proper and effective treatments. Conventional treatments of diabetic LEAD include lifestyle management, medication, open surgery, endovascular treatment, and amputation. As interdisciplinary research emerges, regenerative medicine strategies have provided new insights to treat chronic limb threatening ischemia (CLTI). Therapeutic angiogenesis strategies, such as delivering growth factors, stem cells, drugs to ischemic tissues, have also been proposed to treat LEAD by fundamentally stimulating multidimensional vascular regeneration. Recent years have seen the rapid growth of tissue engineering technology; tissue-engineered biomaterials have been used to study the treatment of LEAD, such as encapsulation of growth factors and drugs in hydrogel to facilitate the restoration of blood perfusion in ischemic tissues of animals. The primary purpose of this review is to introduce treatments and novel biomaterials development in LEAD. Firstly, the pathogenesis of LEAD is briefly described. Secondly, conventional therapies and therapeutic angiogenesis strategies of LEAD are discussed. Finally, recent research advances and future perspectives on biomaterials in LEAD are proposed.
Collapse
Affiliation(s)
- Yang Pan
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuting Luo
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Hong
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huacheng He
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang, China
- *Correspondence: Huacheng He, ; Hong Zhu,
| | - Lu Dai
- The Fourth Outpatient Department, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hong Zhu
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- *Correspondence: Huacheng He, ; Hong Zhu,
| | - Jiang Wu
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
7
|
Nakafusa Y, Nitta N, Ishii K, Shirasu N, Iwamoto T, Nemoto T, Nakamura M, Goto M, Iwata H, Taniguchi M, Yasunami Y. Acceptance of Murine Islet Allografts Without Immunosuppression in Inguinal Subcutaneous White Adipose Tissue Pretreated With bFGF. Diabetes 2022; 71:1721-1734. [PMID: 35604856 DOI: 10.2337/db21-0684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 04/24/2022] [Indexed: 11/13/2022]
Abstract
Prevention of immune rejection without immunosuppression is the ultimate goal of transplant immunobiology. One way to achieve this in cellular transplantation, such as with islet transplantation, is to create a favorable local environment at the transplant site. In the current study, we found that C57BL/6 mice with streptozotocin-induced diabetes remained normoglycemic for >1 year after transplantation of BALB/c islets without immunosuppression when the inguinal subcutaneous white adipose tissue (ISWAT) was the site of transplantation and when the site was pretreated with basic fibroblast growth factor. Mechanistically, mesenchymal stem cells (MSCs) expanded in the ISWAT after the treatment was found to produce transforming growth factor-β (TGF-β), and prevention of islet allograft rejection could be achieved by cotransplantation with syngeneic MSCs isolated from the ISWAT after the treatment, which was abolished by anti-TGF-β antibody treatment. Importantly, TGF-β-producing cells remained present at the site of cotransplantation up to the end of observation period at 240 days after transplantation. These findings indicate that prevention of islet allograft rejection without immunosuppression is feasible with the use of syngeneic TGF-β-producing MSCs expanded in the ISWAT after the treatment with bFGF, providing a novel strategy for prevention of islet allograft rejection without immunosuppression.
Collapse
Affiliation(s)
- Yuki Nakafusa
- Research Institute for Islet Biology, Fukuoka University Central Research Organization, Fukuoka, Japan
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoyoshi Nitta
- Research Institute for Islet Biology, Fukuoka University Central Research Organization, Fukuoka, Japan
| | - Kazunari Ishii
- Department of Microbiology and Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Naoto Shirasu
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Takahiro Iwamoto
- Research Institute for Islet Biology, Fukuoka University Central Research Organization, Fukuoka, Japan
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Takayuki Nemoto
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Hiroo Iwata
- Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, Japan
| | - Masaru Taniguchi
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Yohichi Yasunami
- Research Institute for Islet Biology, Fukuoka University Central Research Organization, Fukuoka, Japan
| |
Collapse
|
8
|
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: 0.3] [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
|
9
|
Plicosepalus acacia Extract and Its Major Constituents, Methyl Gallate and Quercetin, Potentiate Therapeutic Angiogenesis in Diabetic Hind Limb Ischemia: HPTLC Quantification and LC-MS/MS Metabolic Profiling. Antioxidants (Basel) 2021; 10:antiox10111701. [PMID: 34829572 PMCID: PMC8614836 DOI: 10.3390/antiox10111701] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 11/23/2022] Open
Abstract
Plicosepalus acacia (Fam. Loranthaceae) has been reported to possess hypoglycemic, antioxidant, antimicrobial, and anti-inflammatory effects. Liquid chromatography combined with tandem mass spectrometry (LC-MS/MS) analysis revealed the presence of a high content of polyphenolic compounds that are attributed to the therapeutic effects of the crude extract. In addition, methyl gallate and quercetin were detected as major phytomedicinal agents at concentrations of 1.7% and 0.062 g%, respectively, using high-performance thin layer chromatography (HPTLC). The present study investigated the effect of the P. acacia extract and its isolated compounds, methyl gallate and quercetin, on hind limb ischemia induced in type 1 diabetic rats. Histopathological examination revealed that treatment with P. acacia extract, methyl gallate, and quercetin decreased degenerative changes and inflammation in the ischemic muscle. Further biochemical assessment of the hind limb tissue showed decreased oxidative stress, increased levels of nitric oxide and endothelial nitric oxide synthase (eNOS), and enhancement of the levels of heme oxygenase-1 (HO-1) and vascular endothelial growth factor (VEGF) in the groups treated with methyl gallate and quercetin. Expression levels of hypoxia inducible factor-1 alpha (HIF-1α), VEGF, fibroblast growth factor-2 (FGF-2), and miR-146a were upregulated in the muscle tissue of methyl gallate- and quercetin-treated groups along with downregulation of nuclear factor kappa B (NF-κB). In conclusion, P. acacia extract and its isolated compounds, methyl gallate and quercetin, mediated therapeutic angiogenesis in diabetic hind limb ischemia.
Collapse
|
10
|
Dhall S, Park MS, Li C, Sathyamoorthy M. Regenerative Effects of Hypoxia Primed Flowable Placental Formulation in Muscle and Dermal Injury. Int J Mol Sci 2021; 22:7151. [PMID: 34281205 PMCID: PMC8267721 DOI: 10.3390/ijms22137151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 11/17/2022] Open
Abstract
The placental tissue, due to its angiogenic, anti-inflammatory, antioxidative, antimicrobial, and anti-fibrotic properties, has become a compelling source towards a solution for several indications in regenerative medicine. However, methods to enhance and capture the therapeutic properties with formulations that can further the applications of viable placental tissue have not been explored. In this study, we investigated the regenerative effects of a hypoxia primed flowable placental formulation (FPF), composed of amnion/chorion and umbilical tissue, in two in vivo injury models. Laser Doppler data from rodent ischemia hindlimbs treated with FPF revealed significant tissue perfusion improvements compared to control ischemic hindlimbs. To further corroborate FPF's effects, we used a rodent ischemic bipedicle skin flap wound model. FPF treatment significantly increased the rate of wound closure and the quality of wound healing. FPF-treated wounds displayed reduced inflammation and an increase in angiogenesis. Furthermore, quantitative PCR and next-generation sequencing analysis confirmed these changes in the FPF-treated group at both the gene and transcriptional level. The observed modulation in miRNAs was associated with angiogenesis, regulation of inflammatory microenvironment, cell migration and apoptosis, reactive oxygen species generation, and restoring epithelial barrier function, all processes involved in impaired tissue healing. Taken together, these data validate the tissue regenerative properties of the flowable placental formulation configuration tested.
Collapse
Affiliation(s)
- Sandeep Dhall
- Smith & Nephew Plc., Columbia, MD 21046, USA; (C.L.); (M.S.)
| | - Min Sung Park
- Smith & Nephew Plc., Columbia, MD 21046, USA; (C.L.); (M.S.)
| | | | | |
Collapse
|
11
|
Huang SJ, Lee SY, Teng YH, Lee SD, Cheng YJ. Photobiomodulation Therapy to Promote Angiogenesis in Diabetic Mice with Hindlimb Ischemia. PHOTOBIOMODULATION PHOTOMEDICINE AND LASER SURGERY 2021; 39:453-462. [PMID: 34264768 DOI: 10.1089/photob.2020.4896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective: To assess whether photobiomodulation therapy (PBMT) induces angiogenesis in diabetic mice with hindlimb ischemia (HLI). Background: Patients with diabetes mellitus (DM) are at high risk of developing peripheral arterial disease (PAD) in the lower extremities. PBMT has been shown to promote angiogenesis both in vitro and in vivo and could be a treatment for DM patients with PAD. Methods: Femoral artery ligation/excision in mice was performed to induce HLI as an animal model of PAD. PBMT at a dose of 660 nm and 1.91 J/cm2 was delivered for 10 min on 5 consecutive days after the HLI surgery. Control mice received HLI only. Mice in the DM group were injected with streptozocin to induce diabetes before HLI surgery. Mice in the laser and DM+ laser groups received both HLI and PBMT, and the latter group had induced DM. After the laser treatment, lower limb blood flow was evaluated by laser Doppler. The capillary density and CD31 were analyzed by immunofluorescence staining, and protein levels of vascular endothelial growth factor (VEGF)-A, hypoxia-inducible factor-1α (HIF-1α), inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS), and extracellular signal-regulated kinases (ERK) were measured by Western blotting of tissue samples. Results: Compared with the control and DM mice, the laser and DM+ laser groups had more than double the capillary density and blood perfusion rate. Levels of CD31 and VEGF-A proteins in groups that received laser were increased by 1.9- to 3.2-fold compared with groups that did not undergo laser treatment. Animals treated with PBMT exhibited significantly increased HIF-1α expression and ERK phosphorylation compared with animals that did not receive this treatment, and the amount of phospho-eNOS and iNOS increased and decreased, respectively. Conclusions: PBMT can induce therapeutic angiogenesis, indicating that low intensity laser could be a novel treatment for PAD patients.
Collapse
Affiliation(s)
- Shi-Jie Huang
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
| | - Shin-Yi Lee
- General Education Center, China Medical University, Taichung, Taiwan
| | - Yi-Hsien Teng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
| | - Shin-Da Lee
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
| | - Yu-Jung Cheng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan.,Department of Rehabilitation, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
12
|
Nakano T, Mizumasa T, Kuroki Y, Eriguchi M, Yoshida H, Taniguchi M, Masutani K, Tsuruya K, Kitazono T. Advanced glycation end products are associated with immature angiogenesis and peritoneal dysfunction in patients on peritoneal dialysis. Perit Dial Int 2021; 40:67-75. [PMID: 32063152 DOI: 10.1177/0896860819878344] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Deposition of advanced glycation end products (AGEs) is frequently found in the peritoneum of patients on peritoneal dialysis (PD). Angiogenesis is also observed in the peritoneum. However, the clinical significance of AGEs and angiogenesis in the peritoneum is not fully understood. We evaluated the maturation of capillary vessels and investigated whether AGEs are associated with angiogenesis and peritoneal function in the peritoneal membrane. METHODS Peritoneum obtained when PD catheters were removed from 61 patients with PD was analyzed. The peritoneum was immunohistochemically stained with anti-CD34 (for endothelial cells), anti-alpha smooth muscle actin (αSMA) (for pericytes), and anti-AGE antibodies. We defined CD34-positive and αSMA-negative vessels as immature capillary vessels in peritoneal membranes using serial sections. We evaluated the associations between vessel density, peritoneal function (dialysate-to-plasma ratio for creatinine (D/P creatinine)), and the degree of AGE deposition. RESULTS AGE accumulation in the interstitium was positively associated with the duration of PD (p < 0.01). AGE accumulation in the interstitium and vascular wall was positively correlated with the use of acidic solution (p < 0.05) and the maximum value of D/P creatinine (p < 0.05). AGE accumulation in the vascular wall was significantly associated with immature capillary density (CD34+/αSMA-) in the peritoneum (p < 0.01). Vessel density was not significantly correlated with the last measurement of D/P creatinine (p = 0.126, r = 0.202), However, immature capillary density was positively correlated with the last measurement of D/P creatinine (p < 0.05, r = 0.278). CONCLUSIONS AGE accumulation is significantly associated with immature angiogenesis and peritoneal dysfunction in patients undergoing PD.
Collapse
Affiliation(s)
- Toshiaki Nakano
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tohru Mizumasa
- Department of Nephrology, Fukuoka Red Cross Hospital, Japan.,Department of Nephrology, Kyushu Central Hospital, Fukuoka, Japan
| | - Yusuke Kuroki
- Department of Nephrology, Fukuoka Red Cross Hospital, Japan
| | - Masahiro Eriguchi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hisako Yoshida
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatomo Taniguchi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosuke Masutani
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhiko Tsuruya
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Nephrology, Nara Medical University, Kashihara, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
13
|
Yamaki Y, Fukushima T, Yoshida N, Nishimura K, Fukuda A, Hisatake K, Aso M, Sakasai T, Kijima-Tanaka J, Miwa Y, Nakanishi M, Sumazaki R, Takada H. Utilization of a novel Sendai virus vector in ex vivo gene therapy for hemophilia A. Int J Hematol 2021; 113:493-499. [PMID: 33385293 DOI: 10.1007/s12185-020-03059-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 01/19/2023]
Abstract
Sendai virus (SeV) vectors are being recognized as a superior tool for gene transfer. Here, we report the transfection efficacy of a novel, high-performance, replication-defective, and persistent Sendai virus (SeVdp) vector in cultured cells and in mice using a near-infrared fluorescent protein (iRFP)-mediated in vivo imaging system. The novel SeVdp vector established persistent infection, and strong expression of inserted genes was sustained indefinitely in vitro. Analysis of iRFP-expressing cells transplanted subcutaneously into NOG, nude, and ICR mice suggests that innate immunity was involved in the exclusion of the transplanted cells. We also evaluated the feasibility of this novel SeVdp vector for hemophilia A gene therapy. This system enabled insertion of full-length FVIII genes, and transduced cells secreted FVIII into the culture medium. Transient FVIII activity was detected in the plasma of mice after intraperitoneal transplantation of these FVIII-secreting cells. Further improvement in methods to evade immunity, such as simultaneous expression of immunomodulatory genes, would make this novel vector a very useful tool in regenerative medicine.
Collapse
Affiliation(s)
- Yuni Yamaki
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan.
| | - Takashi Fukushima
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan.,Department of Child Health, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Department of Pediatric Tumor, Saitama Medical University International Medical Center, Saitama, Japan
| | - Naomi Yoshida
- Biotechnology Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Ibaraki, Japan.,TOKIWA-Bio Inc., Ibaraki, Japan
| | - Ken Nishimura
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Aya Fukuda
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Koji Hisatake
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | | | - Tomoki Sakasai
- Department of Molecular Pharmacology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Junko Kijima-Tanaka
- Department of Molecular Pharmacology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yoshihiro Miwa
- Department of Molecular Pharmacology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Mahito Nakanishi
- Biotechnology Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Ibaraki, Japan.,TOKIWA-Bio Inc., Ibaraki, Japan
| | - Ryo Sumazaki
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Ibaraki Children's Hospital, Ibaraki, Japan
| | - Hidetoshi Takada
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 305-8576, Japan.,Department of Child Health, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
14
|
Thummarati P, Kino-Oka M. Effect of Co-culturing Fibroblasts in Human Skeletal Muscle Cell Sheet on Angiogenic Cytokine Balance and Angiogenesis. Front Bioeng Biotechnol 2020; 8:578140. [PMID: 33072729 PMCID: PMC7542332 DOI: 10.3389/fbioe.2020.578140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/01/2020] [Indexed: 12/26/2022] Open
Abstract
Skeletal muscle comprises a heterogeneous population of myoblasts and fibroblasts. Autologous skeletal muscle myoblasts are transplanted to patients with ischemia to promote cardiac regeneration. In damaged hearts, various cytokines secreted from the skeletal muscle myoblasts promote angiogenesis and consequently the recovery of cardiac functions. However, the effect of skeletal muscle fibroblasts co-cultured with skeletal muscle myoblasts on angiogenic cytokine production and angiogenesis has not been fully understood. To investigate these effects, production of vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) was measured using the culture medium of monolayers prepared from various cell densities (mono-culture) and proportions (co-culture) of human skeletal muscle myoblasts (HSMMs) and human skeletal muscle fibroblasts (HSMFs). HSMM and HSMF mono-cultures produced VEGF, whereas HSMF mono-culture produced HGF. The VEGF productivity observed in a monolayer comprising low proportion of HSMFs was two-fold greater than that of HSMM and HSMF mono-cultures. The production of VEGF in HSMMs but not in HSMFs was directly proportional to the cell density. VEGF productivity in non-confluent cells with low cell-to-cell contact was higher than that in confluent cells with high cell-to-cell contact. The dynamic migration of cells in a monolayer was examined to analyze the effect of HSMFs on myoblast-to-myoblast contact. The random and rapid migration of HSMFs affected the directional migration of surrounding HSMMs, which disrupted the myoblast alignment. The effect of heterogeneous populations of skeletal muscle cells on angiogenesis was evaluated using human umbilical vein endothelial cells (HUVECs) incubated with fabricated multilayer HSMM sheets comprising various proportions of HSMFs. Co-culturing HSMFs in HSMM sheet at suitable ratio (30 or 40%) enhances endothelial network formation. These findings indicate the role of HSMFs in maintaining cytokine balance and consequently promoting angiogenesis in the skeletal muscle cell sheets. This approach can be used to improve transplantation efficiency of engineered tissues.
Collapse
Affiliation(s)
- Parichut Thummarati
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| |
Collapse
|
15
|
Lee JJ, Arpino JM, Yin H, Nong Z, Szpakowski A, Hashi AA, Chevalier J, O'Neil C, Pickering JG. Systematic Interrogation of Angiogenesis in the Ischemic Mouse Hind Limb: Vulnerabilities and Quality Assurance. Arterioscler Thromb Vasc Biol 2020; 40:2454-2467. [PMID: 32787524 PMCID: PMC7505144 DOI: 10.1161/atvbaha.120.315028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: There has been little success in translating preclinical studies of mouse hind limb ischemia into benefit for patients with peripheral artery disease. Using systematic strategies, we sought to define the injury and angiogenesis landscapes in mice subjected to hind limb ischemia and ascertain whether published studies to date have used an analysis strategy concordant with these data. Approach and Results: Maps of ischemic injury were generated from 22 different hind limb muscles and 33 muscle territories in 12-week-old C57BL/6 mice, based on loss or centralization of myofiber nuclei. Angiogenesis was similarly mapped based on CD (cluster of differentiation) 31–positive capillary content. Only 10 of 33 muscle territories displayed consistent muscle injury, with the distal anterior hind limb muscles most reliably injured. Angiogenesis was patchy and exclusively associated with zones of regenerated muscle (central nuclei). Angiogenesis was not observed in normal appearing muscle, necrotic muscle, or injury border zones. Systematic review of mouse hind limb angiogenesis studies identified 5147 unique publications, of which 509 met eligibility criteria for analysis. Only 7% of these analyzed manuscripts evaluated angiogenesis in distal anterior hind limb muscles and only 15% consistently examined for angiogenesis in zones of muscle regeneration. Conclusions: In 12-week C57BL/6 mice, angiogenesis postfemoral artery excision proceeds exclusively in zones of muscle regeneration. Only a minority of studies to date have analyzed angiogenesis in regions of demonstrably regenerating muscle or in high-likelihood territories. Quality assurance standards, informed by the atlas and mapping data herein, could augment data reliability and potentially help translate mouse hind limb ischemia studies to patient care.
Collapse
Affiliation(s)
- Jason J Lee
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada.,Department of Medicine (J.J.L., A.A.H., J.G.P.), Western University, London, Ontario, Canada.,Department of Medical Biophysics (J.J.L., J.-M.A., J.C., J.G.P.), Western University, London, Ontario, Canada
| | - John-Michael Arpino
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada.,Department of Medical Biophysics (J.J.L., J.-M.A., J.C., J.G.P.), Western University, London, Ontario, Canada
| | - Hao Yin
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada
| | - Zengxuan Nong
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada
| | - Alexis Szpakowski
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada
| | - Abdulaziz A Hashi
- Department of Medicine (J.J.L., A.A.H., J.G.P.), Western University, London, Ontario, Canada
| | - Jacqueline Chevalier
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada.,Department of Medical Biophysics (J.J.L., J.-M.A., J.C., J.G.P.), Western University, London, Ontario, Canada
| | - Caroline O'Neil
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada
| | - J Geoffrey Pickering
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada.,Department of Medicine (J.J.L., A.A.H., J.G.P.), Western University, London, Ontario, Canada.,Department of Medical Biophysics (J.J.L., J.-M.A., J.C., J.G.P.), Western University, London, Ontario, Canada.,Department of Biochemistry (J.G.P.), Western University, London, Ontario, Canada
| |
Collapse
|
16
|
Jiang G, Xiao G, Luo C, Tang Z, Teng Z, Peng X. Correlation Between SNPs at the 3'UTR of the FGF2 Gene and Their Interaction with Environmental Factors in Han Chinese Diabetic Peripheral Neuropathy Patients. J Mol Neurosci 2020; 71:203-214. [PMID: 32613556 DOI: 10.1007/s12031-020-01641-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 06/19/2020] [Indexed: 12/12/2022]
Abstract
FGF2 is a neurotrophic factor that can act as a key regulatory molecule of neuroprotection, neurogenesis, and angiogenesis in various injuries. To explore the genetic background of the FGF2 gene on DPN development, this study analyzed the correlation between SNPs in the 3'UTR of the FGF2 gene and their interaction with environmental factors in DPN patients of Han Chinese nationality. Sanger sequencing was used to analyze the FGF2 genotypes at the rs1048201, rs3804158, rs41348645, rs6854081, rs3747676, rs7683093, rs1476215, and rs1476217 loci in 150 DPN patients, 150 NDPN patients, and 150 healthy control patients. Plasma FGF2 levels were measured in all subjects by using ELISAs. Subjects carrying the T allele at the rs1048201 locus in the FGF2 gene had a significantly lower risk of developing DPN compared with subjects carrying the C allele (OR = 0.43, 95% CI = 0.33-0.56, p < 0.01). Subjects with the G genotype at the rs6854081 locus had an exceptionally higher risk of developing DPN than subjects with the T allele (OR = 1.66, 95% CI = 1.39-1.89, p < 0.01). Individuals harboring the G allele at the rs7683093 locus had a markedly higher risk of DPN than patients with the C allele (OR = 1.63, 95% CI = 1.36-1.87, p < 0.01). Finally, individuals having the A genotype at the rs1476215 locus had a significantly higher risk of DPN than individuals carrying the T allele (OR = 1.82, 95% CI = 1.53-2.02, p < 0.01). There was an interaction between age and alcohol consumption and the SNP rs7683093. SNPs at rs1048201, rs6854081, rs7683093, and rs1476215 in the FGF2 3'UTR were strongly associated with plasma levels of FGF2 (p < 0.05). SNPs at the rs1048201, rs6854081, rs7683093, and rs1476215 loci in the FGF2 gene were significantly associated with the risk of DPN. A possible mechanism is that these SNPs affect the expression level of FGF2 by interrupting the binding of microRNAs to target sites in the 3'UTR.
Collapse
Affiliation(s)
- Guangyuan Jiang
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Gang Xiao
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Chao Luo
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Zhaohua Tang
- Departmen of neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhipeng Teng
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xing Peng
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China.
| |
Collapse
|
17
|
Yao Z, Liu H, Yang M, Bai Y, Zhang B, Wang C, Yan Z, Niu G, Zou Y, Li Y. Bone marrow mesenchymal stem cell-derived endothelial cells increase capillary density and accelerate angiogenesis in mouse hindlimb ischemia model. Stem Cell Res Ther 2020; 11:221. [PMID: 32513272 PMCID: PMC7278145 DOI: 10.1186/s13287-020-01710-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/20/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) can improve limb perfusion and increase vessel density in a murine model of hindlimb ischemia. But low engraftment rate of those cells limited their therapeutic effect. Endothelial cells (ECs) play an important role in neovascularization. And MSCs can differentiate into ECs in vitro. The aim of this study is to investigate if EC differentiation of MSCs in vitro before transplantation is effective in improving therapeutic outcomes in the treatment of ischemic disease in a murine ischemia animal model. Methods MSCs were isolated from the bone marrow of EGFP-transgenic mice by density gradient centrifugation. The identity of the MSCs was determined by their cluster of differentiation (CD) marker profile by flow cytometry. Inducing medium containing a few cytokines was applied to induce the MSCs to differentiate into ECs. Endothelial differentiation was quantitatively evaluated using flow cytometry, quantitative real-time PCR (qRT-PCR), immunofluorescence, Matrigel tube formation assay, and Dil-labeled acetylated low-density lipoprotein uptake assay. Mouse hindlimb ischemia model was made by excision of the femoral artery. Uninduced EGFP+ MSCs, induced EGFP+ MSCs, and PBS were intramuscularly injected into the gastrocnemius following ischemia no later than 24 h after operation. Restoration of blood flow and muscle function was evaluated by laser Doppler perfusion imaging. Immunofluorescence was conducted to evaluate the engraftment of transplanted MSCs. Histological analysis was performed to evaluate blood vessel formation. Results Induced EGFP+ MSCs expressed endothelial markers and exhibited tube formation capacity. Mice in the induced EGFP+ MSCs group had a better blood perfusion recovery, enhanced vessel densities, higher engraftment, and improved function of the ischemic limb than those in the uninduced EGFP+ MSCs or PBS groups. Conclusions This study reveals that after short-term pre-treatment in the EC-inducing medium, induced MSCs acquire stronger vessel formation capability and enhanced angiogenic therapeutic effect in the murine hindlimb ischemia model.
Collapse
Affiliation(s)
- Ziping Yao
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Huihui Liu
- Department of Hematology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Min Yang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Yun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Bihui Zhang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Chengen Wang
- Department of Minimally Invasive Tumor Therapies Center, National Center of Gerontology, Beijing Hospital, Beijing, China
| | - Ziguang Yan
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Guochen Niu
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Yinghua Zou
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China.
| | - Yuan Li
- Department of Hematology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China.
| |
Collapse
|
18
|
Kokubu S, Inaki R, Hoshi K, Hikita A. Adipose-derived stem cells improve tendon repair and prevent ectopic ossification in tendinopathy by inhibiting inflammation and inducing neovascularization in the early stage of tendon healing. Regen Ther 2020; 14:103-110. [PMID: 31989000 PMCID: PMC6970144 DOI: 10.1016/j.reth.2019.12.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Achilles tendinopathy is characterized by scar formation or ectopic ossification, both of which result in pain and worsened physical function in athletes and older people. Although cell therapy using adipose-derived stem cells (ASCs) has been shown to be effective for tendinopathy, the underlying mechanisms by which ASCs result in tendon healing in vivo have not yet been fully clarified. METHODS ASCs were obtained from the fat pads of EGFP transgenic mice by collagenase digestion. C57BL/6 mice were used in a collagenase-induced injury model. ASCs were transplanted into injury sites at 1 week after injury. Tendons were harvested at 9 days, 2 weeks, and 4 weeks after transplantation, and analyzed by histological examination and μCT scanning. RESULTS Histological analysis and μCT scanning revealed greater recovery of collagen fibers and suppression of ectopic ossification in the ASC-treated group than in the control group at 2 and 4 weeks after injury. Immunohistochemical analysis identified transplanted ASCs in the tendon core close to peritenon and connective tissue at 2 days and 1 week after transplantation, but not at 3 weeks. Furthermore, while the expression levels of IL-1β, GLUT1, and CA9 were significantly reduced in the ASC group compared to the control group at 9 days after injury, those of VEGF and the number of CD31 positive vessels were significantly increased. CONCLUSION The efficacy of ASCs for tendon repair and the prevention of ectopic ossification in Achilles tendinopathy were demonstrated. Our data suggest that ASCs can modulate inflammation and induce neovascularization in the early stage of tendon injury.
Collapse
Affiliation(s)
- Saeko Kokubu
- Department of Sensory and Motor System Medicine, Department of Oral and Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ryoko Inaki
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazuto Hoshi
- Department of Oral-maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Atsuhiko Hikita
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
19
|
Murai Y, Sasase T, Tadaki H, Heitaku S, Imagawa N, Yamada T, Ohta T. Analysis of haemodynamics and angiogenic response to ischaemia in the obese type 2 diabetic model Spontaneously Diabetic Torii
Lepr
fa
(SDT fatty) rats. Clin Exp Pharmacol Physiol 2020; 47:583-590. [DOI: 10.1111/1440-1681.13239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/14/2019] [Accepted: 12/15/2019] [Indexed: 01/14/2023]
Affiliation(s)
- Yasutaka Murai
- Japan Tobacco Inc. Central Pharmaceutical Research Institute Takatsuki Osaka Japan
- Faculty of Agriculture Department of Agrobiology Niigata University Niigata Japan
| | - Tomohiko Sasase
- Japan Tobacco Inc. Central Pharmaceutical Research Institute Takatsuki Osaka Japan
| | - Hironobu Tadaki
- Japan Tobacco Inc. Central Pharmaceutical Research Institute Takatsuki Osaka Japan
| | - Shiro Heitaku
- Japan Tobacco Inc. Central Pharmaceutical Research Institute Takatsuki Osaka Japan
| | - Naoya Imagawa
- Japan Tobacco Inc. Central Pharmaceutical Research Institute Takatsuki Osaka Japan
| | - Takahisa Yamada
- Faculty of Agriculture Department of Agrobiology Niigata University Niigata Japan
| | - Takeshi Ohta
- Laboratory of Animal Physiology and Functional Anatomy Graduate School of Agriculture Kyoto University Kitashirakawa Kyoto Japan
| |
Collapse
|
20
|
Abstract
Myocardial ischemia and peripheral vascular disease persist as significant clinical problems despite improved medical, surgical, and endovascular therapies. Advances in our understanding of the biological mechanisms that govern capillary neovascularization and collateral artery growth have enabled molecular therapies for revascularizing ischemic tissues. Generally known as therapeutic angiogenesis, this review summarizes the essential pre-clinical research and the major clinical trials of molecular therapies for ischemic disease. Early clinical experience has established the proof of principle, however, inconsistent and modest improvements in clinical outcomes have exposed the complexity of neovascularization and problems with transitioning basic science to clinical applicability.
Collapse
Affiliation(s)
- Shant M Vartanian
- Division of Vascular Surgery, University of California, San Francisco, California 94143-0104, USA.
| | | |
Collapse
|
21
|
Hassanshahi M, Khabbazi S, Peymanfar Y, Hassanshahi A, Hosseini-Khah Z, Su YW, Xian CJ. Critical limb ischemia: Current and novel therapeutic strategies. J Cell Physiol 2019; 234:14445-14459. [PMID: 30637723 DOI: 10.1002/jcp.28141] [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] [Received: 11/25/2018] [Accepted: 01/02/2019] [Indexed: 01/24/2023]
Abstract
Critical limb ischemia (CLI) is the advanced stage of peripheral artery disease spectrum and is defined by limb pain or impending limb loss because of compromised blood flow to the affected extremity. Current conventional therapies for CLI include amputation, bypass surgery, endovascular therapy, and pharmacological approaches. Although these conventional therapeutic strategies still remain as the mainstay of treatments for CLI, novel and promising therapeutic approaches such as proangiogenic gene/protein therapies and stem cell-based therapies have emerged to overcome, at least partially, the limitations and disadvantages of current conventional therapeutic approaches. Such novel CLI treatment options may become even more effective when other complementary approaches such as utilizing proper bioscaffolds are used to increase the survival and engraftment of delivered genes and stem cells. Therefore, herein, we address the benefits and disadvantages of current therapeutic strategies for CLI treatment and summarize the novel and promising therapeutic approaches for CLI treatment. Our analyses also suggest that these novel CLI therapeutic strategies show considerable advantages to be used when current conventional methods have failed for CLI treatment.
Collapse
Affiliation(s)
- Mohammadhossein Hassanshahi
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Samira Khabbazi
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Yaser Peymanfar
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Alireza Hassanshahi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Zahra Hosseini-Khah
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Yu-Wen Su
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Cory J Xian
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
22
|
Abstract
The ability to generate new microvessels in desired numbers and at desired locations has been a long-sought goal in vascular medicine, engineering, and biology. Historically, the need to revascularize ischemic tissues nonsurgically (so-called therapeutic vascularization) served as the main driving force for the development of new methods of vascular growth. More recently, vascularization of engineered tissues and the generation of vascularized microphysiological systems have provided additional targets for these methods, and have required adaptation of therapeutic vascularization to biomaterial scaffolds and to microscale devices. Three complementary strategies have been investigated to engineer microvasculature: angiogenesis (the sprouting of existing vessels), vasculogenesis (the coalescence of adult or progenitor cells into vessels), and microfluidics (the vascularization of scaffolds that possess the open geometry of microvascular networks). Over the past several decades, vascularization techniques have grown tremendously in sophistication, from the crude implantation of arteries into myocardial tunnels by Vineberg in the 1940s, to the current use of micropatterning techniques to control the exact shape and placement of vessels within a scaffold. This review provides a broad historical view of methods to engineer the microvasculature, and offers a common framework for organizing and analyzing the numerous studies in this area of tissue engineering and regenerative medicine. © 2019 American Physiological Society. Compr Physiol 9:1155-1212, 2019.
Collapse
Affiliation(s)
- Joe Tien
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Division of Materials Science and Engineering, Boston University, Brookline, Massachusetts, USA
| |
Collapse
|
23
|
Okadome J, Matsumoto T, Yoshiya K, Matsuda D, Tamada K, Onimaru M, Nakano K, Egashira K, Yonemitsu Y, Maehara Y. BubR1 insufficiency impairs angiogenesis in aging and in experimental critical limb ischemic mice. J Vasc Surg 2018; 68:576-586.e1. [DOI: 10.1016/j.jvs.2017.07.119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/10/2017] [Indexed: 01/01/2023]
|
24
|
Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R, Augustin HG, Bates DO, van Beijnum JR, Bender RHF, Bergers G, Bikfalvi A, Bischoff J, Böck BC, Brooks PC, Bussolino F, Cakir B, Carmeliet P, Castranova D, Cimpean AM, Cleaver O, Coukos G, Davis GE, De Palma M, Dimberg A, Dings RPM, Djonov V, Dudley AC, Dufton NP, Fendt SM, Ferrara N, Fruttiger M, Fukumura D, Ghesquière B, Gong Y, Griffin RJ, Harris AL, Hughes CCW, Hultgren NW, Iruela-Arispe ML, Irving M, Jain RK, Kalluri R, Kalucka J, Kerbel RS, Kitajewski J, Klaassen I, Kleinmann HK, Koolwijk P, Kuczynski E, Kwak BR, Marien K, Melero-Martin JM, Munn LL, Nicosia RF, Noel A, Nurro J, Olsson AK, Petrova TV, Pietras K, Pili R, Pollard JW, Post MJ, Quax PHA, Rabinovich GA, Raica M, Randi AM, Ribatti D, Ruegg C, Schlingemann RO, Schulte-Merker S, Smith LEH, Song JW, Stacker SA, Stalin J, Stratman AN, Van de Velde M, van Hinsbergh VWM, Vermeulen PB, Waltenberger J, Weinstein BM, Xin H, Yetkin-Arik B, Yla-Herttuala S, Yoder MC, Griffioen AW. Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 2018; 21:425-532. [PMID: 29766399 PMCID: PMC6237663 DOI: 10.1007/s10456-018-9613-x] [Citation(s) in RCA: 435] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, University of Lausanne, Rue Michel-Servet 1, CMU, 1211, Geneva 4, Switzerland.
- Translational Research Center in Oncohaematology, University of Geneva, Geneva, Switzerland.
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Elizabeth Allen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
| | - Andrey Anisimov
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Alfred C Aplin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Hellmut G Augustin
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - David O Bates
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - R Hugh F Bender
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Andreas Bikfalvi
- Angiogenesis and Tumor Microenvironment Laboratory (INSERM U1029), University Bordeaux, Pessac, France
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Barbara C Böck
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - Peter C Brooks
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Federico Bussolino
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute-FPO-IRCCS, 10060, Candiolo, Italy
| | - Bertan Cakir
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anca M Cimpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - George Coukos
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine and Dalton Cardiovascular Center, Columbia, MO, USA
| | - Michele De Palma
- School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Emily Couric Cancer Center, The University of Virginia, Charlottesville, VA, USA
| | - Neil P Dufton
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute, Leuven, Belgium
| | | | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, UK
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bart Ghesquière
- Metabolomics Expertise Center, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, Metabolomics Expertise Center, KU Leuven, Leuven, Belgium
| | - Yan Gong
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Adrian L Harris
- Molecular Oncology Laboratories, Oxford University Department of Oncology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Nan W Hultgren
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | | | - Melita Irving
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Robert S Kerbel
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois, Chicago, IL, USA
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hynda K Kleinmann
- The George Washington University School of Medicine, Washington, DC, USA
| | - Pieter Koolwijk
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Elisabeth Kuczynski
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Juan M Melero-Martin
- Department of Cardiac Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roberto F Nicosia
- Department of Pathology, University of Washington, Seattle, WA, USA
- Pathology and Laboratory Medicine Service, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Agnes Noel
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Jussi Nurro
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Tatiana V Petrova
- Department of oncology UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund, Sweden
| | - Roberto Pili
- Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Jeffrey W Pollard
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Mark J Post
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Department Surgery, LUMC, Leiden, The Netherlands
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, National Council of Scientific and Technical Investigations (CONICET), Buenos Aires, Argentina
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Anna M Randi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
- National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Curzio Ruegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Steven A Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre and The Sir Peter MacCallum, Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Jimmy Stalin
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Amber N Stratman
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Maureen Van de Velde
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Victor W M van Hinsbergh
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium
- Translational Cancer Research Unit, GZA Hospitals, Sint-Augustinus & University of Antwerp, Antwerp, Belgium
| | - Johannes Waltenberger
- Medical Faculty, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hong Xin
- University of California, San Diego, La Jolla, CA, USA
| | - Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Seppo Yla-Herttuala
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Del Giudice C, Ifergan G, Goudot G, Bellamy V, Messas E, Clement O, Bruneval P, Menasche P, Sapoval M. Evaluation of a new model of hind limb ischemia in rabbits. J Vasc Surg 2017; 68:849-857. [PMID: 29074110 DOI: 10.1016/j.jvs.2017.07.140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/28/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Various animal models of critical limb ischemia have been developed in the past. However, there is no animal model that can undergo endovascular treatment, while providing reproducible true critical limb ischemia with arterial ulcers and rest pain. We evaluated the efficacy of a new model of rabbit hindlimb ischemia created through a percutaneous approach using embolization with calibrated particles. METHODS Through a percutaneous transauricular artery approach and selective catheterization of the superficial femoral artery, embolization of distal limb vessels was performed using a mixture of 300- to 500-μm calibrated microparticles (Embosphere, Merit Medical, Salt Lake City, Utah), saline solution, and iodine contrast. Clinical and ultrasound imaging-based blood flow evaluation was performed before embolization and during follow-up. Histologic evaluation was performed at humane killing 14 days after the procedure. RESULTS The model was successfully created in 10 rabbits (10 limbs). One rabbit died of sudden death at 8 days after the procedure. The nine surviving rabbits developed hind ulcers. All rabbits had a higher pain score in the follow-up compared to baseline value (P < .0001). Blood flow in the saphenous artery decreased significantly after the procedure and later at 14 days follow-up (baseline value 63.4 ± 31.3 μL per cardiac cycle vs 32.0 ± 28.4 μL per cardiac cycle postprocedure [P = .0013] and 32.0 ± 28.4 μL per cardiac cycle at 14 days [P = .0015]). Pathology showed signs of severe limb ischemia in all rabbits with subacute and chronic injury patterns. CONCLUSIONS A rabbit hind limb ischemia model created by percutaneous transauricular distal femoral artery embolization with calibrated particles may overcome some of the limitations of existing animal models. As such, this model could prove useful for assessing therapies designed to improve arterial perfusion and collateral growth.
Collapse
Affiliation(s)
- Costantino Del Giudice
- Vascular and Oncological Interventional Radiology, Université Paris Descartes, Hôpital Européen George Pompidou, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| | - Gabriel Ifergan
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Guillaume Goudot
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Angiology, Université Paris Descartes, Paris, France
| | - Valerie Bellamy
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Emmanuel Messas
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Angiology, Université Paris Descartes, Paris, France
| | - Olivier Clement
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Department of Radiology, Université Paris Descartes, Paris, France
| | - Patrick Bruneval
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Department of Pathology, Université Paris Descartes, Hôpital Européen George Pompidou, Paris, France
| | - Philippe Menasche
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Department of Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Marc Sapoval
- Vascular and Oncological Interventional Radiology, Université Paris Descartes, Hôpital Européen George Pompidou, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
26
|
A novel strategy to enhance angiogenesis in vivo using the small VEGF-binding peptide PR1P. Angiogenesis 2017; 20:399-408. [PMID: 28397127 DOI: 10.1007/s10456-017-9556-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/04/2017] [Indexed: 12/21/2022]
Abstract
Therapeutic angiogenesis is an experimental frontier in vascular biology that seeks to deliver angiogenic growth factors to ischemic or injured tissues to promote targeted formation of new blood vessels as an alternative approach to surgical revascularization procedures. Vascular endothelial growth factor (VEGF) is a potent angiogenic signal protein that is locally upregulated at sites of tissue injury. However, therapies aimed at increasing VEGF levels experimentally by injecting VEGF gene or protein failed to improve outcomes in human trials in part due to its short half-life and systemic toxicity. We recently designed a novel 12-amino acid peptide (PR1P) whose sequence was derived from an extracellular VEGF-binding domain of the pro-angiogenic glycoprotein prominin-1. In this study, we characterized the molecular binding properties of this novel potential therapeutic for targeted angiogenesis and provided the foundation for its use as an angiogenic molecule that can potentiate endogenous VEGF. We showed that PR1P bound VEGF directly and enhanced VEGF binding to endothelial cells and to VEGF receptors VEGFR2 and neuropilin-1. PR1P increased angiogenesis in the murine corneal micropocket assay when combined with VEGF, but had no activity without added VEGF. In addition, PR1P also enhanced angiogenesis in murine choroidal neovascularization and wound-healing models and augmented reperfusion in a murine hind-limb ischemia model. Together our data suggest that PR1P enhanced angiogenesis by potentiating the activity of endogenous VEGF. In so doing, this novel therapy takes advantage of endogenous VEGF gradients generated in injured tissues and may improve the efficacy of and avoid systemic toxicity seen with previous VEGF therapies.
Collapse
|
27
|
Chloroquine improves the response to ischemic muscle injury and increases HMGB1 after arterial ligation. J Vasc Surg 2017; 67:910-921. [PMID: 28259568 DOI: 10.1016/j.jvs.2017.01.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/09/2017] [Indexed: 12/30/2022]
Abstract
OBJECTIVE We have previously shown that exogenous administration of the nuclear protein high mobility group box 1 (HMGB1) improves angiogenesis after tissue ischemia. Antagonizing HMGB1 prolongs muscle necrosis and deters regeneration. In this study, we evaluated HMGB1 expression in peripheral arterial disease (PAD) and the mechanisms that promote its release in a murine model of hindlimb ischemia. Specifically, we investigated how chloroquine (CQ), a commonly employed disease-modifying antirheumatic drug, promotes HMGB1 release from muscle. We hypothesized that CQ could increase HMGB1 locally and systemically, allowing it to mediate recovery from ischemic injury. METHODS Muscle biopsies were performed on patients undergoing lower extremity surgery for non-PAD-related disease as well as for claudication and critical limb ischemia. Clinical symptoms and ankle-brachial indices were recorded for each patient. HMGB1 was detected in muscle sections using immunohistochemical staining. Unilateral femoral artery ligation was performed on both wild-type and inducible HMGB1 knockout mice. Wild-type mice were administered intraperitoneal CQ 2 weeks before and after femoral artery ligation. Laser Doppler perfusion imaging was used to determine perfusion recovery. Serum and tissue levels of HMGB1 were measured at designated time points. In vitro, cultured C2C12 myoblasts were treated with increasing doses of CQ. HMGB1, autophagosome formation, p62/SQSTM1 accumulation, caspase-1 expression and activity, and lactate dehydrogenase levels were measured in supernatants and cell lysates. RESULTS Nuclear expression of HMGB1 was prominent in patients with claudication and critical limb ischemia (P < .05) compared with controls. CQ-treated mice had elevated serum HMGB1 and diffuse HMGB1 staining in muscle (P < .01). In wild-type mice, CQ treatment resulted in higher laser Doppler perfusion imaging ratios in the ischemic limb at 7 days (P < .03) and less fat replacement after 2 weeks (P < .03). In cultured myoblasts, CQ induced autophagosome accumulation, inhibited p62/SQSTM-1 degradation, and activated caspase-1. CONCLUSIONS HMGB1 is prominently expressed in PAD muscle but mostly confined to the nucleus. Our in vivo data suggest that HMGB1 mobilization into the sarcoplasm and serum can be increased with CQ, possibly through caspase-1-mediated pathways. Whereas HMGB1 can be released by many cell types, these studies suggest that the muscle may be an important additional source that is relevant in PAD.
Collapse
|
28
|
Hsieh MJ, Liu HT, Wang CN, Huang HY, Lin Y, Ko YS, Wang JS, Chang VHS, Pang JHS. Therapeutic potential of pro-angiogenic BPC157 is associated with VEGFR2 activation and up-regulation. J Mol Med (Berl) 2016; 95:323-333. [PMID: 27847966 DOI: 10.1007/s00109-016-1488-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/20/2016] [Accepted: 11/07/2016] [Indexed: 02/06/2023]
Abstract
BPC 157, a pentadecapeptide with extensive healing effects, has recently been suggested to contribute to angiogenesis. However, the underlying mechanism is not yet clear. The present study aimed to explore the potential therapeutic effect and pro-angiogenic mechanism of BPC 157. As demonstrated by the chick chorioallantoic membrane (CAM) assay and endothelial tube formation assay, BPC 157 could increase the vessel density both in vivo and in vitro, respectively. BPC 157 could also accelerate the recovery of blood flow in the ischemic muscle of the rat hind limb as detected by laser Doppler scanning, indicating the promotion of angiogenesis. Histological analysis of the hind limb muscle confirmed the increased number of vessels and the enhanced vascular expression of vascular endothelial growth factor receptor 2 (VEGFR2) in rat with BPC 157 treatment. In vitro study using human vascular endothelial cells further confirmed the increased mRNA and protein expressions of VEGFR2 but not VEGF-A by BPC 157. In addition, BPC 157 could promote VEGFR2 internalization in vascular endothelial cells which was blocked in the presence of dynasore, an inhibitor of endocytosis. BPC 157 time dependently activated the VEGFR2-Akt-eNOS signaling pathway which could also be suppressed by dynasore. The increase of endothelial tube formation induced by BPC 157 was also inhibited by dynasore. This study demonstrates the pro-angiogenic effects of BPC 157 that is associated with the increased expression, internalization of VEGFR2, and the activation of VEGFR2-Akt-eNOS signaling pathway. BPC 157 promotes angiogenesis in CAM assay and tube formation assay. BPC 157 accelerates the blood flow recovery and vessel number in rats with hind limb ischemia. BPC 157 up-regulates VEGFR2 expression in rats with hind limb ischemia and endothelial cell culture. BPC 157 promotes VEGFR2 internalization in association with VEGFR2-Akt-eNOS activation. KEY MESSAGE BPC 157 promotes angiogenesis in CAM assay and tube formation assay. BPC 157 accelerates the blood flow recovery and vessel number in rats with hind limb ischemia. BPC 157 up-regulates VEGFR2 expression in rats with hind limb ischemia and endothelial cell culture. BPC 157 promotes VEGFR2 internalization in association with VEGFR2-Akt-eNOS activation.
Collapse
Affiliation(s)
- Ming-Jer Hsieh
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan City, Taiwan, Republic Of China.,Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Lin-kou, Chang Gung University, Tao-Yuan City, Taiwan, Republic Of China
| | - Hsien-Ta Liu
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan City, Taiwan, Republic Of China.,Division of Family Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, Republic Of China.,School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chao-Nin Wang
- Department of Obstetrics and Gynecology, Lin-Kou Medical Center, Chang Gung Memorial Hospital, Tao-Yuan City, Taiwan, Republic Of China
| | - Hsiu-Yun Huang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan City, Taiwan, Republic Of China
| | - Yuling Lin
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan City, Taiwan, Republic Of China
| | - Yu-Shien Ko
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Lin-kou, Chang Gung University, Tao-Yuan City, Taiwan, Republic Of China
| | - Jong-Shyan Wang
- Healthy Aging Research Center, Graduate Institute of Rehabilitation Science, Medical College, Chang Gung University, Tao-Yuan City, Taiwan, Republic Of China
| | - Vincent Hung-Shu Chang
- Program for Translation Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, Republic Of China
| | - Jong-Hwei S Pang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan City, Taiwan, Republic Of China. .,Department of Physical Medicine and Rehabilitation, Lin-Kou Medical Center, Chang Gung Memorial Hospital, Tao-Yuan City, Taiwan, Republic Of China.
| |
Collapse
|
29
|
Jazwa A, Florczyk U, Grochot-Przeczek A, Krist B, Loboda A, Jozkowicz A, Dulak J. Limb ischemia and vessel regeneration: Is there a role for VEGF? Vascul Pharmacol 2016; 86:18-30. [PMID: 27620809 DOI: 10.1016/j.vph.2016.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/24/2016] [Accepted: 09/07/2016] [Indexed: 11/18/2022]
Abstract
Vascular endothelial growth factor (VEGF), as an endothelial cell-specific mitogen, is crucial for new blood vessels formation. Atherosclerosis affecting the cardiovascular system causes ischemia and functio laesa in tissues supplied by the occluded vessels. When such a situation occurs in the lower extremities, it causes critical limb ischemia (CLI) often requiring leg amputation. Low oxygen tension leads to upregulation of hypoxia-regulated genes (i.e. VEGF), that should help to restore the impaired blood flow. In CLI these rescue mechanisms are, however, often inefficient. Moreover, there are many contradictory reports showing either induction, no changes or even down-regulation of VEGF in specimens taken from patients with CLI, as well as in samples collected from animals subjected to hindlimb ischemia. Additionally, taking into account numerous experimental and clinical data demonstrating rather insufficient therapeutic potential of VEGF, we called into question the role of this protein in limb ischemia and vessel regeneration. In this review we are also summarizing several aspects which can influence VEGF expression and its measurement in the ischemic tissues.
Collapse
Affiliation(s)
- Agnieszka Jazwa
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Urszula Florczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Bart Krist
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
30
|
Hwang PTJ, Lim DJ, Fee T, Alexander GC, Tambralli A, Andukuri A, Tian L, Cui W, Berry J, Gilbert SR, Jun HW. A bio-inspired hybrid nanosack for graft vascularization at the omentum. Acta Biomater 2016; 41:224-34. [PMID: 27286678 PMCID: PMC4969099 DOI: 10.1016/j.actbio.2016.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/31/2016] [Accepted: 06/07/2016] [Indexed: 01/16/2023]
Abstract
UNLABELLED For three-dimensional tissue engineering scaffolds, the major challenges of hydrogels are poor mechanical integrity and difficulty in handling during implantation. In contrast, electrospun scaffolds provide tunable mechanical properties and high porosity; but, are limited in cell encapsulation. To overcome these limitations, we developed a "hybrid nanosack" by combination of a peptide amphiphile (PA) nanomatrix gel and an electrospun poly (ε-caprolactone) (ePCL) nanofiber sheet with porous crater-like structures. This hybrid nanosack design synergistically possessed the characteristics of both approaches. In this study, the hybrid nanosack was applied to enhance local angiogenesis in the omentum, which is required of tissue engineering scaffolds for graft survival. The ePCL sheet with porous crater-like structures improved cell and blood vessel penetration through the hybrid nanosack. The hybrid nanosack also provided multi-stage fibroblast growth factor-2 (FGF-2) release kinetics for stimulating local angiogenesis. The hybrid nanosack was implanted into rat omentum for 14days and vascularization was analyzed by micro-CT and immunohistochemistry; the data clearly demonstrated that both FGF-2 delivery and porous crater-like structures work synergistically to enhance blood vessel formation within the hybrid nanosack. Therefore, the hybrid nanosack will provide a new strategy for engineering scaffolds to achieve graft survival in the omentum by stimulating local vascularization, thus overcoming the limitations of current strategies. STATEMENT OF SIGNIFICANCE For three-dimensional tissue engineering scaffolds, the major challenges of hydrogels are poor mechanical integrity and difficulty in handling during implantation. In contrast, electrospun scaffolds provide tunable mechanical properties and high porosity; but, are limited in cell encapsulation. To overcome these limitations, we developed a "hybrid nanosack" by combination of a peptide amphiphile (PA) nanomatrix gel and an electrospun poly (ε-caprolactone) (ePCL) nanofiber sheet with porous crater-like structures. This design synergistically possessed the characteristics of both approaches. In this study, the hybrid nanosack was applied to enhance local angiogenesis in the omentum, which is required of tissue engineering scaffolds for graft survival. The hybrid nanosack was implanted into rat omentum for 14days and vascularization was analyzed by micro-CT and immunohistochemistry. We demonstrate that both FGF-2 delivery and porous crater-like structures work synergistically to enhance blood vessel formation within the hybrid nanosack. Therefore, the hybrid nanosack will provide a new strategy for engineering scaffolds to achieve graft survival in the omentum by stimulating local vascularization, thus overcoming the limitations of current strategies.
Collapse
Affiliation(s)
- Patrick T J Hwang
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Dong-Jin Lim
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy Fee
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Grant C Alexander
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Ajay Tambralli
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Adinarayana Andukuri
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Liqun Tian
- Department of Surgery, University of Alabama at Birmingham, AL 35294, USA
| | - Wanxing Cui
- Medstar Georgetown Transplant Institute, Medstar Georgetown University Hospital, Washington DC 20007, USA
| | - Joel Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Shawn R Gilbert
- Department of Surgery, University of Alabama at Birmingham, AL 35294, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA.
| |
Collapse
|
31
|
Matsumoto T, Tanaka M, Yoshiya K, Yoshiga R, Matsubara Y, Horiuchi-Yoshida K, Yonemitsu Y, Maehara Y. Improved quality of life in patients with no-option critical limb ischemia undergoing gene therapy with DVC1-0101. Sci Rep 2016; 6:30035. [PMID: 27418463 PMCID: PMC4945920 DOI: 10.1038/srep30035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/29/2016] [Indexed: 11/09/2022] Open
Abstract
Critical limb ischemia (CLI) has a poor prognosis and adversely affects patients' quality of life (QOL). Therapeutic angiogenesis may improve mobility, mortality, and QOL in CLI patients. However, the effectiveness of gene therapy on such patients' QOL is unknown. DVC1-0101, a non-transmissible recombinant Sendai virus vector expressing human fibroblast growth factor-2 gene, demonstrated safety and efficacy in a phase I/II study of CLI patients. We investigated the effects of DVC1-0101 on QOL in this cohort. QOL was assessed using the Short Form-36 health survey version 2 (SF-36) in 12 patients at pre-administration, 28 days, and 3, 6, and 12 months post-treatment. We examined differences between pre and post-administration QOL scores and correlations between QOL scores and vascular parameters. Patients demonstrated low baselines scores on every SF-36 dimension. Post-treatment scores showed significant improvements in physical functioning at 3 and 6 months (P < 0.05), role-physical at 3, 6, and 12 months (P < 0.05), bodily pain at 1, 3, 6, and 12 months (P < 0.05), vitality at 1, 6, and 12 months (P < 0.05), and physical component summary at 6 and 12 months (P < 0.05). DVC1-0101-based gene therapy may improve QOL in CLI patients over a 6-month period.
Collapse
Affiliation(s)
- Takuya Matsumoto
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Michiko Tanaka
- R&D Laboratory for Innovative Biotherapeutics Science, Graduate School of Pharmaceutical Sciences, Kyushu University, Rm. 601, Collaborative Research Station I, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Keiji Yoshiya
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ryosuke Yoshiga
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yutaka Matsubara
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kumi Horiuchi-Yoshida
- R&D Laboratory for Innovative Biotherapeutics Science, Graduate School of Pharmaceutical Sciences, Kyushu University, Rm. 601, Collaborative Research Station I, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshikazu Yonemitsu
- R&D Laboratory for Innovative Biotherapeutics Science, Graduate School of Pharmaceutical Sciences, Kyushu University, Rm. 601, Collaborative Research Station I, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
32
|
Ye L, Haider HK, Jiang SJ, Sim EKW. Therapeutic Angiogenesis Using Vascular Endothelial Growth Factor. Asian Cardiovasc Thorac Ann 2016; 12:173-81. [PMID: 15213090 DOI: 10.1177/021849230401200221] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Therapeutic angiogenesis using vascular endothelial growth factor can reduce tissue ischemia by simulating the natural process of angiogenesis. Vascular endothelial growth factor not only stimulates endothelial cells to proliferate and migrate, but also mobilizes endothelial progenitor cells and achieves vascular protection. Besides direct administration of angiogenic proteins, plasmids and viral vectors carrying angiogenic genes have been used. Animal experiments have shown promise with evidence of neovascularization and improved perfusion in the target myocardium. Initial phase I and II clinical trials results are encouraging and reflect the potential success of therapeutic angiogenesis as a clinical modality for the treatment of ischemic heart disease. This review discusses the role of vascular endothelial growth factor in therapeutic angiogenesis, along with the problems and considerations of this approach as a treatment strategy.
Collapse
Affiliation(s)
- Lei Ye
- Department of Cardiothoracic and Vascular Surgery, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
33
|
El-Sakka AI. What is the current role of intracavernosal injection in management of erectile dysfunction? Int J Impot Res 2016; 28:88-95. [DOI: 10.1038/ijir.2016.14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 11/14/2015] [Accepted: 03/14/2016] [Indexed: 12/15/2022]
|
34
|
Kim JH, Lee HJ, Song YS. Mesenchymal stem cell-based gene therapy for erectile dysfunction. Int J Impot Res 2016; 28:81-7. [PMID: 26888355 DOI: 10.1038/ijir.2016.3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 10/17/2015] [Accepted: 11/23/2015] [Indexed: 12/12/2022]
Abstract
Despite the overwhelming success of PDE5 inhibitor (PDE5I), the demand for novel pharmacotherapeutic and surgical options for ED continues to rise owing to the increased proportion of elderly individuals in the population, in addition to the growing percentage of ED patients who do not respond to PDE5I. Surgical treatment of ED is associated with many complications, thus warranting the need for nonsurgical therapies. Moreover, none of the above-mentioned treatments essentially corrects, cures or prevents ED. Although gene therapy is a promising option, many challenges and obstacles such as local inflammatory response and random transgene expression, in addition to other safety issues, limit its use at the clinical level. The use of stem cell therapy alone also has many shortcomings. To overcome these inadequacies, many scientists and clinicians are investigating new gene and stem cell therapies.
Collapse
Affiliation(s)
- J H Kim
- Department of Urology, Soonchunhyang University Hospital, College of Medicine, Soonchunhyang University, Seoul, Korea
| | - H J Lee
- Biomedical Research Institute, Chung-Ang School of Medicine, Seoul, Korea
| | - Y S Song
- Department of Urology, Soonchunhyang University Hospital, College of Medicine, Soonchunhyang University, Seoul, Korea
| |
Collapse
|
35
|
Rioja AY, Tiruvannamalai Annamalai R, Paris S, Putnam AJ, Stegemann JP. Endothelial sprouting and network formation in collagen- and fibrin-based modular microbeads. Acta Biomater 2016; 29:33-41. [PMID: 26481042 PMCID: PMC4681647 DOI: 10.1016/j.actbio.2015.10.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/04/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022]
Abstract
A modular tissue engineering approach may have advantages over current therapies in providing rapid and sustained revascularization of ischemic tissue. In this study, modular protein microbeads were prepared from pure fibrin (FIB) and collagen-fibrin composites (COL-FIB) using a simple water-in-oil emulsification technique. Human endothelial cells and fibroblasts were embedded directly in the microbead matrix. The resulting microbeads were generally spheroidal with a diameter of 100-200μm. Cell viability was high (75-80% viable) in microbeads, but was marginally lower than in bulk hydrogels of corresponding composition (85-90% viable). Cell proliferation was significantly greater in COL-FIB microbeads after two weeks in culture, compared to pure FIB microbeads. Upon embedding of microbeads in a surrounding fibrin hydrogel, endothelial cell networks formed inside the microbead matrix and extended into the surrounding matrix. The number of vessel segments, average segment length, and number of branch points was higher in FIB samples, compared to COL-FIB samples, resulting in significantly longer total vessel networks. Anastomosis of vessel networks from adjacent microbeads was also observed. These studies demonstrate that primitive vessel networks can be formed by modular protein microbeads containing embedded endothelial cells and fibroblasts. Such microbeads may find utility as prevascularized tissue modules that can be delivered minimally invasively as a therapy to restore blood flow to ischemic tissues. STATEMENT OF SIGNIFICANCE Vascularization is critically important for tissue engineering and regenerative medicine, and materials that support and/or promote neovascularization are of value both for translational applications and for mechanistic studies and discovery-based research. Therefore, we fabricated small modular microbeads formulated from pure fibrin (FIB) and collagen-fibrin (COL-FIB) containing endothelial cells and supportive fibroblasts. We explored how cells encapsulated within these materials form microvessel-like networks both within and outside of the microbeads when embedded in larger 3D matrices. FIB microbeads were found to initiate more extensive sprouting into the surrounding ECM in vitro. These results represent an important step towards our goal of developing injectable biomaterial modules containing preformed vascular units that can rapidly restore vascularization to an ischemic tissue in vivo.
Collapse
Affiliation(s)
- Ana Y Rioja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States
| | | | - Spencer Paris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States
| | - Andrew J Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States.
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States.
| |
Collapse
|
36
|
Evaluation of the clinical relevance and limitations of current pre-clinical models of peripheral artery disease. Clin Sci (Lond) 2015; 130:127-50. [DOI: 10.1042/cs20150435] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peripheral artery disease (PAD) has recognized treatment deficiencies requiring the discovery of novel interventions. This article describes current animal models of PAD and discusses their advantages and disadvantages. There is a need for models which more directly simulate the characteristics of human PAD, such as acute-on-chronic presentation, presence of established risk factors and impairment of physical activity.
Collapse
|
37
|
Li S, Dang Y, Zhou X, Huang B, Huang X, Zhang Z, Kwan YW, Chan SW, Leung GPH, Lee SMY, Hoi MPM. Formononetin promotes angiogenesis through the estrogen receptor alpha-enhanced ROCK pathway. Sci Rep 2015; 5:16815. [PMID: 26568398 PMCID: PMC4645220 DOI: 10.1038/srep16815] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/14/2015] [Indexed: 11/09/2022] Open
Abstract
Formononetin is an isoflavone that has been shown to display estrogenic properties and induce angiogenesis activities. However, the interrelationship between the estrogenic properties and angiogenesis activities of formononetin are not well defined. In the present study, docking and enzymatic assay demonstrated that formononetin displayed direct binding to the ligand-binding domain (LBD) of estrogen receptor alpha (ERα) with an agonistic property. Results from Human Umbilical Vein Endothelial Cells (HUVEC) by using real-time migration xCELLigence system, immunofluorescence and western blotting provided strong evidences of formononetin induced endothelial cell migration and dramatic actin cytoskeleton spatial modification through ERα-enhanced-ROCK-II/MMP2/9 signaling pathways. In addition, results from co-immunoprecipitation suggested formononetin induced cell migration via recruiting of ERα/ROCK-II activated complex formation. More interestingly, in zebrafish embryo we observed that formononetin significantly promoted angiogenic sproutings in the subintestinal vessels (SIVs) that could be completely abolished by ROCK inhibitor. In this study, we elucidated the underlying mechanisms that formononetin produced proangiogenesis effects through an ERα-enhanced ROCK-II signaling pathways. Results from the present study also expand our knowledge about the enigmatic underlying mechanisms of phytoestrogenic compounds in the promotion of angiogenesis in relation to ERα and ROCK interaction in endothelial cells and their relationship with actin assembly and cell migration.
Collapse
Affiliation(s)
- Shang Li
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yuanye Dang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xuelin Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Bin Huang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xiaohui Huang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zherui Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yiu Wa Kwan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Shun Wan Chan
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - George Pak Heng Leung
- Pharmacology and Pharmacy, Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Simon Ming Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
38
|
Zheng LZ, Cao HJ, Chen SH, Tang T, Fu WM, Huang L, Chow DHK, Wang YX, Griffith JF, He W, Zhou H, Zhao DW, Zhang G, Wang XL, Qin L. Blockage of Src by Specific siRNA as a Novel Therapeutic Strategy to Prevent Destructive Repair in Steroid-Associated Osteonecrosis in Rabbits. J Bone Miner Res 2015; 30:2044-57. [PMID: 25917347 DOI: 10.1002/jbmr.2542] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 04/21/2015] [Accepted: 04/22/2015] [Indexed: 12/15/2022]
Abstract
Vascular hyperpermeability and highly upregulated bone resorption in the destructive repair progress of steroid-associated osteonecrosis (SAON) are associated with a high expression of VEGF and high Src activity (Src is encoded by the cellular sarcoma [c-src] gene). This study was designed to prove our hypothesis that blocking the VEGF-Src signaling pathway by specific Src siRNA is able to prevent destructive repair in a SAON rabbit model. Destructive repair in SAON was induced in rabbits. At 2, 4, and 6 weeks after SAON induction, VEGF, anti-VEGF, Src siRNA, Src siRNA+VEGF, control siRNA, and saline were introduced via intramedullary injection into proximal femora for each group, respectively. Vascularization and permeability were quantified by dynamic contrast-enhanced (DCE) MRI. At week 6 after SAON induction, proximal femurs were dissected for micro-computed tomography (μCT)-based trabecular architecture with finite element analysis (FEA), μCT-based angiography, and histological analysis. Histological evaluation revealed that VEGF enhanced destructive repair, whereas anti-VEGF prevented destructive repair and Src siRNA and Src siRNA+VEGF prevented destructive repair and enhanced reparative osteogenesis. Findings of angiography and histomorphometry were consistent with those determined by DCE MRI. Src siRNA inhibited VEGF-mediated vascular hyperpermeability but preserved VEGF-induced neovascularization. Bone resorption was enhanced in the VEGF group and inhibited in the anti-VEGF, Src siRNA, Src siRNA+VEGF groups as determined by both 3D μCT and 2D histomorphometry. FEA showed higher estimated failure load in the Src siRNA and Src siRNA+VEGF groups when compared to the vehicle control group. Blockage of VEGF-Src signaling pathway by specific Src siRNA was able to prevent steroid-associated destructive repair while improving reconstructive repair in SAON, which might become a novel therapeutic strategy.
Collapse
Affiliation(s)
- Li-zhen Zheng
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Hui-juan Cao
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Shi-hui Chen
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Tao Tang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.,Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Wei-min Fu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.,Department of Orthopaedics, Zhongshan Hospital of Dalian University, Dalian, PR China
| | - Le Huang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Yi-xiang Wang
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - James Francis Griffith
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Wei He
- Department of Orthopaedics, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, PR China
| | - Hong Zhou
- Bone Research Program, ANZAC (Australian and New Zealand Army Corps.) Research Institute, University of Sydney, Sydney, Australia
| | - De-wei Zhao
- Department of Orthopaedics, Zhongshan Hospital of Dalian University, Dalian, PR China
| | - Ge Zhang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Xin-luan Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.,Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.,Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| |
Collapse
|
39
|
Heuslein JL, Meisner JK, Li X, Song J, Vincentelli H, Leiphart RJ, Ames EG, Blackman BR, Blackman BR, Price RJ. Mechanisms of Amplified Arteriogenesis in Collateral Artery Segments Exposed to Reversed Flow Direction. Arterioscler Thromb Vasc Biol 2015; 35:2354-65. [PMID: 26338297 PMCID: PMC4618717 DOI: 10.1161/atvbaha.115.305775] [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: 04/20/2015] [Accepted: 08/14/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Collateral arteriogenesis, the growth of existing arterial vessels to a larger diameter, is a fundamental adaptive response that is often critical for the perfusion and survival of tissues downstream of chronic arterial occlusion(s). Shear stress regulates arteriogenesis; however, the arteriogenic significance of reversed flow direction, occurring in numerous collateral artery segments after femoral artery ligation, is unknown. Our objective was to determine if reversed flow direction in collateral artery segments differentially regulates endothelial cell signaling and arteriogenesis. APPROACH AND RESULTS Collateral segments experiencing reversed flow direction after femoral artery ligation in C57BL/6 mice exhibit increased pericollateral macrophage recruitment, amplified arteriogenesis (30% diameter and 2.8-fold conductance increases), and remarkably permanent (12 weeks post femoral artery ligation) remodeling. Genome-wide transcriptional analyses on human umbilical vein endothelial cells exposed to reversed flow conditions mimicking those occurring in vivo yielded 10-fold more significantly regulated transcripts, as well as enhanced activation of upstream regulators (nuclear factor κB [NFκB], vascular endothelial growth factor, fibroblast growth factor-2, and transforming growth factor-β) and arteriogenic canonical pathways (protein kinase A, phosphodiesterase, and mitogen-activated protein kinase). Augmented expression of key proarteriogenic molecules (Kruppel-like factor 2 [KLF2], intercellular adhesion molecule 1, and endothelial nitric oxide synthase) was also verified by quantitative real-time polymerase chain reaction, leading us to test whether intercellular adhesion molecule 1 or endothelial nitric oxide synthase regulate amplified arteriogenesis in flow-reversed collateral segments in vivo. Interestingly, enhanced pericollateral macrophage recruitment and amplified arteriogenesis was attenuated in flow-reversed collateral segments after femoral artery ligation in intercellular adhesion molecule 1(-/-) mice; however, endothelial nitric oxide synthase(-/-) mice showed no such differences. CONCLUSIONS Reversed flow leads to a broad amplification of proarteriogenic endothelial signaling and a sustained intercellular adhesion molecule 1-dependent augmentation of arteriogenesis. Further investigation of the endothelial mechanotransduction pathways activated by reversed flow may lead to more effective and durable therapeutic options for arterial occlusive diseases.
Collapse
Affiliation(s)
- Joshua L Heuslein
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Joshua K Meisner
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Xuanyue Li
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Ji Song
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Helena Vincentelli
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Ryan J Leiphart
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Elizabeth G Ames
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Brett R Blackman
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | | | - Richard J Price
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.).
| |
Collapse
|
40
|
Kurioka T, Mizutari K, Niwa K, Fukumori T, Inoue M, Hasegawa M, Shiotani A. Hyaluronic acid pretreatment for Sendai virus-mediated cochlear gene transfer. Gene Ther 2015; 23:187-95. [PMID: 26361273 DOI: 10.1038/gt.2015.94] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/19/2015] [Accepted: 09/04/2015] [Indexed: 12/14/2022]
Abstract
Gene therapy with viral vectors is one of the most promising strategies for sensorineural hearing loss. However, safe and effective administration of the viral vector into cochlear tissue is difficult because of the anatomical isolation of the cochlea. We investigated the efficiency and safety of round window membrane (RWM) application of Sendai virus, one of the most promising non-genotoxic vectors, after pretreatment with hyaluronic acid (HA) on the RWM to promote efficient viral translocation into the cochlea. Sendai virus expressing the green fluorescent protein reporter gene was detected throughout cochlear tissues following application combined with HA pretreatment. Quantitative analysis revealed that maximum expression was reached 3 days after treatment. The efficiency of transgene expression was several 100-fold greater with HA pretreatment than that without. Furthermore, unlike the conventional intracochlear delivery methods, this approach did not cause hearing loss. These findings reveal the potential utility of gene therapy with Sendai virus and HA for treatment of sensorineural hearing loss.
Collapse
Affiliation(s)
- T Kurioka
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - K Mizutari
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - K Niwa
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - T Fukumori
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - M Inoue
- DNAVEC Corporation, Ibaraki, Japan
| | | | - A Shiotani
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| |
Collapse
|
41
|
Kang HM, Sohn I, Jung J, Jeong JW, Park C. Exendin-4 protects hindlimb ischemic injury by inducing angiogenesis. Biochem Biophys Res Commun 2015; 465:758-63. [PMID: 26299927 DOI: 10.1016/j.bbrc.2015.08.080] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/18/2015] [Indexed: 11/19/2022]
Abstract
Exendin-4, an analog of glucagon-like peptide-1, has shown to have beneficial effects on endothelial function, and was recently approved for the treatment of diabetes. In previous studies, we showed that exendin-4 induces angiogenesis in in vitro and ex vivo assays; in this study, we assessed the proangiogenic effects of exendin-4 in vivo using a mouse hindlimb ischemia model. Treatment with exendin-4 for three days mitigated hindlimb and gastrocnemius muscle fiber necrosis. Hindlimb perfusion was determined using indocyanine green fluorescence dynamics that showed, significantly higher blood flow rate to the ischemic hindlimbs in an exendin-4-treated group. Immunohistochemistry assay showed that exendin-4 increased CD31-positive areas in the gastrocnemius muscle of ischemic limbs. Furthermore, treatment of the hindlimbs of ischemic mice with exendin-4 increased vascular endothelial growth factor (VEGF) and phospho-extracellular signal-related kinase (ERK) on western blot analysis. Our data demonstrate that exendin-4 prevents hindlimb ischemic injury by inducing vessels via VEGF angiogenic-related pathways. These findings suggest that exendin-4 has potential as a therapeutic agent for vascular diseases that stimulate angiogenesis.
Collapse
Affiliation(s)
- Hye-Min Kang
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea
| | - Inkyung Sohn
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea
| | - Joo-Won Jeong
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea
| | - Chan Park
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea.
| |
Collapse
|
42
|
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: 3.8] [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
|
43
|
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: 13.1] [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
|
44
|
Hoying JB, Utzinger U, Weiss JA. Formation of microvascular networks: role of stromal interactions directing angiogenic growth. Microcirculation 2015; 21:278-89. [PMID: 24447042 PMCID: PMC4032604 DOI: 10.1111/micc.12115] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 01/16/2014] [Indexed: 12/29/2022]
Abstract
In the adult, angiogenesis leads to an expanded microvascular network as new vessel segments are added to an existing microcirculation. Necessarily, growing neovessels must navigate through tissue stroma as they locate and grow toward other vessel elements. We have a growing body of evidence demonstrating that angiogenic neovessels reciprocally interact with the interstitial matrix of the stroma resulting in directed neovascular growth during angiogenesis. Given the compliance and the viscoelastic properties of collagen, neovessel guidance by the stroma is likely due to compressive strain transverse to the direction of primary tensile forces present during active tissue deformation. Similar stromal strains control the final network topology of the new microcirculation, including the distribution of arterioles, capillaries, and venules. In this case, stromal-derived stimuli must be present during the post-angiogenesis remodeling and maturation phases of neovascularization to have this effect. Interestingly, the preexisting organization of vessels prior to the start of angiogenesis has no lasting influence on the final, new network architecture. Combined, the evidence describes interplay between angiogenic neovessels and stroma that is important in directed neovessel growth and invasion. This dynamic is also likely a mechanism by which global tissue forces influence vascular form and function.
Collapse
Affiliation(s)
- James B Hoying
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| | | | | |
Collapse
|
45
|
A new murine model of sustainable and durable chronic critical limb ischemia fairly mimicking human pathology. Eur J Vasc Endovasc Surg 2015; 49:205-12. [PMID: 25579876 DOI: 10.1016/j.ejvs.2014.12.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/04/2014] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To establish a chronic mouse model of critical limb ischemia (CLI) with in vivo and ex vivo validation, closely mimicking human pathology. METHODS Swiss mice (n = 28) were submitted to sequential unilateral femoral (day 0) and iliac (day 4) ligatures. Ischemia was confirmed by clinical scores (tissue and functional damages) and methoxyisobutylisonitrile (MIBI) scintigraphies at days 0, 4, 6, 10, 20, and 30. At days 10, 20, and 30, muscle mitochondrial respiration, calcium retention capacity (CRC), and production of reactive oxygen species (ROS) were investigated, together with transcripts of mitochondrial biogenesis and antioxidant enzymes. Histological analysis was also performed. RESULTS Clinical and functional damage confirmed CLI. MIBI scintigraphies showed hypoperfusion of the ischemic limb, which remained stable until day 30. Mitochondrial respiration was impaired in ischemic muscles compared with controls (Vmax = 7.93 ± 0.99 vs. 10.09 ± 2.87 mmol/L O2/minute/mg dry weight [dw]; p = .01), together with impaired CRC (7.4 ± 1.6 mmol/L minute/mg dw vs. 11.9 ± 0.9 mmol/L minute/mg dw; p < .001) and biogenesis (41% decrease in peroxisome proliferator-activated receptor gamma coactivator [PGC]-1α, 49% decrease in PGC-1β, and 41% decrease in nuclear respiratory factor-1). Ischemic muscles also demonstrated increased production of ROS under electron paramagnetic resonance (0.084 ± 0.029 vs. 0.051 ± 0.031 mmol/L minute/mg dw; p = .03) and with dihydroethidium staining (3622 ± 604 arbitrary units of fluorescence vs. 1224 ± 324; p < .01), decreased antioxidant enzymes (32% decrease in superoxide dismutase [SOD]1, 41% decrease in SOD2, and 49% decrease in catalase), and myopathic features (wider range in fiber size, rounded shape, centrally located nuclei, and smaller cross-sectional areas). All defects were stable over time. CONCLUSION Sequential femoral and iliac ligatures closely mimic human functional, clinical, scintigraphic, and skeletal muscle mitochondrial characteristics, and could prove useful for testing therapeutic approaches.
Collapse
|
46
|
Himeno T, Kamiya H, Naruse K, Cheng Z, Ito S, Shibata T, Kondo M, Kato J, Okawa T, Fujiya A, Suzuki H, Kito T, Hamada Y, Oiso Y, Isobe K, Nakamura J. Angioblast Derived from ES Cells Construct Blood Vessels and Ameliorate Diabetic Polyneuropathy in Mice. J Diabetes Res 2015; 2015:257230. [PMID: 25977928 PMCID: PMC4419216 DOI: 10.1155/2015/257230] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/24/2015] [Accepted: 03/25/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Although numerous reports addressing pathological involvements of diabetic polyneuropathy have been conducted, a universally effective treatment of diabetic polyneuropathy has not yet been established. Recently, regenerative medicine studies in diabetic polyneuropathy using somatic stem/progenitor cell have been reported. However, the effectiveness of these cell transplantations was restricted because of their functional and numerical impairment in diabetic objects. Here, we investigated the efficacy of treatment for diabetic polyneuropathy using angioblast-like cells derived from mouse embryonic stem cells. METHODS AND RESULTS Angioblast-like cells were obtained from mouse embryonic stem cells and transplantation of these cells improved several physiological impairments in diabetic polyneuropathy: hypoalgesia, delayed nerve conduction velocities, and reduced blood flow in sciatic nerve and plantar skin. Furthermore, pathologically, the capillary number to muscle fiber ratios were increased in skeletal muscles of transplanted hindlimbs, and intraepidermal nerve fiber densities were ameliorated in transplanted plantar skin. Transplanted cells maintained their viabilities and differentiated to endothelial cells and smooth muscle cells around the injection sites. Moreover, several transplanted cells constructed chimeric blood vessels with recipient cells. CONCLUSIONS These results suggest that transplantation of angioblast like cells induced from embryonic stem cells appears to be a novel therapeutic strategy for diabetic polyneuropathy.
Collapse
Affiliation(s)
- Tatsuhito Himeno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hideki Kamiya
- Department of Chronic Kidney Disease Initiatives, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 21 Karimata, Yazako, Nagakute, Aichi 480-1195, Japan
- *Hideki Kamiya:
| | - Keiko Naruse
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya 464-8650, Japan
| | - Zhao Cheng
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Sachiko Ito
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Taiga Shibata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masaki Kondo
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Jiro Kato
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tetsuji Okawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Atsushi Fujiya
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
- Department of Metabolic Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hirohiko Suzuki
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tetsutaro Kito
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoji Hamada
- Department of Metabolic Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yutaka Oiso
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kenichi Isobe
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Jiro Nakamura
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 21 Karimata, Yazako, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
47
|
Endothelial cell FGF signaling is required for injury response but not for vascular homeostasis. Proc Natl Acad Sci U S A 2014; 111:13379-84. [PMID: 25139991 DOI: 10.1073/pnas.1324235111] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Endothelial cells (ECs) express fibroblast growth factor receptors (FGFRs) and are exquisitely sensitive to FGF signals. However, whether the EC or another vascular cell type requires FGF signaling during development, homeostasis, and response to injury is not known. Here, we show that Flk1-Cre or Tie2-Cre mediated deletion of FGFR1 and FGFR2 (Fgfr1/2(Flk1-Cre) or Fgfr1/2(Tie2-Cre) mice), which results in deletion in endothelial and hematopoietic cells, is compatible with normal embryonic development. As adults, Fgfr1/2(Flk1-Cre) mice maintain normal blood pressure and vascular reactivity and integrity under homeostatic conditions. However, neovascularization after skin or eye injury was significantly impaired in both Fgfr1/2(Flk1-Cre) and Fgfr1/2(Tie2-Cre) mice, independent of either hematopoietic cell loss of FGFR1/2 or vascular endothelial growth factor receptor 2 (Vegfr2) haploinsufficiency. Also, impaired neovascularization was associated with delayed cutaneous wound healing. These findings reveal a key requirement for cell-autonomous EC FGFR signaling in injury-induced angiogenesis, but not for vascular homeostasis, identifying the EC FGFR signaling pathway as a target for diseases associated with aberrant vascular proliferation, such as age-related macular degeneration, and for modulating wound healing without the potential toxicity associated with direct manipulation of systemic FGF or VEGF activity.
Collapse
|
48
|
|
49
|
Sachdev U, Cui X, Xu J, Xu J, Tzeng E. MyD88 and TRIF mediate divergent inflammatory and regenerative responses to skeletal muscle ischemia. Physiol Rep 2014; 2:2/5/e12006. [PMID: 24844636 PMCID: PMC4098734 DOI: 10.14814/phy2.12006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We have previously shown that MyD88 KO mice appear protected from ischemic muscle injury while TRIF KO mice exhibit sustained necrosis after femoral artery ligation (FAL). However, our previous data did not differentiate whether the protective effect of absent MyD88 signaling was secondary to attenuated injury after FAL or quicker recovery from the insult. The purpose of this study was to delineate these different possibilities. On the basis of previous findings, we hypothesized that MyD88 signaling promotes enhanced inflammation while TRIF mediates regeneration after skeletal muscle ischemia. Our results show that after FAL, both MyD88 KO mice and TRIF KO mice have evidence of ischemia, as do their control counterparts. However, MyD88 KO mice had lower levels of serum IL‐6 24 h after FAL, while TRIF KO mice demonstrated sustained serum IL‐6 up to 1 week after injury. Additionally, MyD88 KO mice had higher nuclear content and larger myofibers than control animals 1 week after injury. IL‐6 is known to have differential effects in myoblast function, and can inhibit proliferation and differentiation. In tibialis anterior muscle harvested from injured animals, IL‐6 levels were higher and the proliferative marker MyoD was lower in TRIF KO mice by PCR. Furthermore, expression of MyD88 appeared to be higher in skeletal muscle of TRIF KO mice. In vitro, we showed that myoblast differentiation and proliferation were attenuated in response to IL‐6 treatment giving credence to the finding that low IL‐6 in MyD88 KO mice may be responsible for larger myocyte sizes 1 week after FAL. We conclude that MyD88 and TRIF work in concert to mediate a balanced response to ischemic injury. We describe opposing roles of MyD88 and TRIF, both downstream signaling molecules of TLR4, in the inflammatory and regenerative processes that follow limb ischemia. MyD88 appears to mediate inflammation, while TRIF appears to be required for modulation of MyD88 activity and promoting regeneration. Absence of MyD88 may ultimately have a protective effect in muscle recovery after ischemic injury.
Collapse
Affiliation(s)
- Ulka Sachdev
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Xiangdong Cui
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jia Xu
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jun Xu
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Edith Tzeng
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania Department of Surgery, VA Pittsburgh Health System, Pittsburgh, Pennsylvania
| |
Collapse
|
50
|
Basu G, Downey H, Guo S, Israel A, Asmar A, Hargrave B, Heller R. Prevention of distal flap necrosis in a rat random skin flap model by gene electrotransfer delivering VEGF165plasmid. J Gene Med 2014; 16:55-65. [DOI: 10.1002/jgm.2759] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Gaurav Basu
- Frank Reidy Research Center for Bioelectrics; Old Dominion University; Norfolk VA USA
| | - Harre Downey
- Frank Reidy Research Center for Bioelectrics; Old Dominion University; Norfolk VA USA
| | - Siqi Guo
- Frank Reidy Research Center for Bioelectrics; Old Dominion University; Norfolk VA USA
| | - Annelise Israel
- Frank Reidy Research Center for Bioelectrics; Old Dominion University; Norfolk VA USA
| | - Anthony Asmar
- Frank Reidy Research Center for Bioelectrics; Old Dominion University; Norfolk VA USA
| | - Barbara Hargrave
- Frank Reidy Research Center for Bioelectrics; Old Dominion University; Norfolk VA USA
- School of Medical Diagnostics and Translational Science; Old Dominion University; Norfolk VA USA
| | - Richard Heller
- Frank Reidy Research Center for Bioelectrics; Old Dominion University; Norfolk VA USA
- School of Medical Diagnostics and Translational Science; Old Dominion University; Norfolk VA USA
| |
Collapse
|