1
|
Jian J, Ji-Qiu C, Zheng-Li C, Shen-Jun S, Lei L, Jie Z, Ming-Yi M, Shi-Jie B, Zai-Fang Z, Shi-Hui Z. Automatic wound closure system: closure of large wounds by stretching the skin around the wound. Updates Surg 2024; 76:2429-2439. [PMID: 39162930 DOI: 10.1007/s13304-024-01850-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/10/2024] [Indexed: 08/21/2024]
Abstract
Large-wound treatment often requires autologous skin grafting or skin flap transfer, causing iatrogenic secondary injuries. Thus, we have developed an automatic wound closure system that consists of a stretch module, microcontroller, and touch screen. Full-thickness wounds (8 × 14 cm) on Bama miniature pigs were manually closed by direct suture in control animals and with three different tension levels performed by the automatic device in the experimental animals. Wound-closure conditions, post-closure healing, and scars were evaluated. Post-operative microscopic changes in collagen fibers, local cell apoptosis, and changes in vascular density were compared between the two wound-closure techniques. In the control group and the first experimental group, which used a traction force of 15 N, primary wound closure could not be achieved. The other two experimental groups used a traction force of 30 N and 60 N and all wounds achieved primary closure. Collagen-fiber stretching was observed histologically in all groups and collagen-fiber breakdown occurred in some wounds when the traction force was 60 N. Scar hyperplasia was significantly reduced in the automatic wound closure system groups. The collagen content decreased, cell apoptosis increased, and vascular density decreased in local tissues in the early post-closure stage, but eventually recovered to normal-skin levels. In summary, we developed an automatic wound closure system that effectively and safely stretches dermal-collagen fibers under an appropriate traction force (30 N) and stretch wound-peripheral skin to cover the wound, achieve primary closure, and reduce scar hyperplasia.
Collapse
Affiliation(s)
- Jin Jian
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, People's Republic of China
- Shanghai Depeac Biotechnology Co., Ltd, Shanghai, People's Republic of China
| | - Chen Ji-Qiu
- Department of Burn Surgery, First Affiliated Hospital of Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Chen Zheng-Li
- Department of Burn Surgery, First Affiliated Hospital of Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Sheng Shen-Jun
- Department of Electromechanical Engineering and Automation, Shanghai University, 99 Shangda Road, Shanghai, 200438, People's Republic of China
| | - Liu Lei
- Department of Burn Surgery, First Affiliated Hospital of Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Zhu Jie
- Shanghai Depeac Biotechnology Co., Ltd, Shanghai, People's Republic of China
| | - Mao Ming-Yi
- Shanghai Depeac Biotechnology Co., Ltd, Shanghai, People's Republic of China
| | - Bai Shi-Jie
- Shanghai Depeac Biotechnology Co., Ltd, Shanghai, People's Republic of China
| | - Zhang Zai-Fang
- Department of Electromechanical Engineering and Automation, Shanghai University, 99 Shangda Road, Shanghai, 200438, People's Republic of China.
| | - Zhu Shi-Hui
- Department of Burn Surgery, First Affiliated Hospital of Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
2
|
Wlodarczyk J, Leng A, Abadchi SN, Shababi N, Mokhtari-Esbuie F, Gheshlaghi S, Ravari MR, Pippenger EK, Afrasiabi A, Ha J, Abraham JM, Harmon JW. Transfection of hypoxia-inducible factor-1α mRNA upregulates the expression of genes encoding angiogenic growth factors. Sci Rep 2024; 14:6738. [PMID: 38509125 PMCID: PMC10954730 DOI: 10.1038/s41598-024-54941-w] [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: 11/08/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
Hypoxia-Inducible Factor-1α (HIF-1α) has presented a new direction for ischemic preconditioning of surgical flaps to promote their survival. In a previous study, we demonstrated the effectiveness of HIF-1a DNA plasmids in this application. In this study, to avoid complications associated with plasmid use, we sought to express HIF-1α through mRNA transfection and determine its biological activity by measuring the upregulation of downstream angiogenic genes. We transfected six different HIF-1a mRNAs-one predominant, three variant, and two novel mutant isoforms-into primary human dermal fibroblasts using Lipofectamine, and assessed mRNA levels using RT-qPCR. At all time points examined after transfection (3, 6, and 10 h), the levels of HIF-1α transcript were significantly higher in all HIF-1α transfected cells relative to the control (all p < 0.05, unpaired Student's T-test). Importantly, the expression of HIF-1α transcription response genes (VEGF, ANG-1, PGF, FLT1, and EDN1) was significantly higher in the cells transfected with all isoforms than with the control at six and/or ten hours post-transfection. All isoforms were transfected successfully into human fibroblast cells, resulting in the rapid upregulation of all five downstream angiogenic targets tested. These findings support the potential use of HIF-1α mRNA for protecting ischemic dermal flaps.
Collapse
Affiliation(s)
- Jakub Wlodarczyk
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
- Department of General and Oncological Surgery, Medical University of Lodz, Lodz, Poland
| | - Albert Leng
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Sanaz Nourmohammadi Abadchi
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Niloufar Shababi
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Farzad Mokhtari-Esbuie
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Shayan Gheshlaghi
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Mohsen Rouhani Ravari
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
- Department of Surgery, University of Chicago Medicine, Chicago, IL, 60637, USA
| | - Emma K Pippenger
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Ali Afrasiabi
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Jinny Ha
- Division of Thoracic Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - John M Abraham
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - John W Harmon
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA.
| |
Collapse
|
3
|
Matsunaga S, Tomita S. [The effect of PHD inhibitor on tumor microenvironment and tumor immune response]. Nihon Yakurigaku Zasshi 2024; 159:169-172. [PMID: 38692882 DOI: 10.1254/fpj.23091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Since the approval of HIF-PH inhibitors, HIF-PH inhibitors have been used clinically, and many studies and clinical case reports have been reported in Japan. A lot of information has been accumulated on clinical usage. However, HIF-PH inhibitors require careful administration for cancer patients due to their action mechanism through upregulating hypoxia-inducible factors (HIFs) level. In cancer cells, HIFs affect tumor progression and contribute to chemo- and radio-resistance. On the other hand, upregulation of HIFs in immune cells is associated with inflammation and suppress tumor progression. However, these controversial effects are not clear in in vivo model. It is needed to reveal whether upregulating HIFs level is beneficial for cancer therapy or not. We have previously reported that HIF-PH inhibitor treatment in tumor bearing mice model led to reconstitute tumor blood vessel and inhibit tumor growth. In addition, these phenomena were caused by tumor infiltrated macrophages and they altered these phenotypes. In this review, we will describe our findings on the mechanism of tumor growth suppression by HIF-PH inhibitors. We also want to mention the risks and benefits of future HIF-PH inhibitors.
Collapse
Affiliation(s)
- Shinji Matsunaga
- Department of Pharmacology, Osaka Metropolitan University Graduate School of Medicine
| | - Shuhei Tomita
- Department of Pharmacology, Osaka Metropolitan University Graduate School of Medicine
| |
Collapse
|
4
|
Shan C, Xia Y, Wu Z, Zhao J. HIF-1α and periodontitis: Novel insights linking host-environment interplay to periodontal phenotypes. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 184:50-78. [PMID: 37769974 DOI: 10.1016/j.pbiomolbio.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/27/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
Periodontitis, the sixth most prevalent epidemic disease globally, profoundly impacts oral aesthetics and masticatory functionality. Hypoxia-inducible factor-1α (HIF-1α), an oxygen-dependent transcriptional activator, has emerged as a pivotal regulator in periodontal tissue and alveolar bone metabolism, exerts critical functions in angiogenesis, erythropoiesis, energy metabolism, and cell fate determination. Numerous essential phenotypes regulated by HIF are intricately associated with bone metabolism in periodontal tissues. Extensive investigations have highlighted the central role of HIF and its downstream target genes and pathways in the coupling of angiogenesis and osteogenesis. Within this concise perspective, we comprehensively review the cellular phenotypic alterations and microenvironmental dynamics linking HIF to periodontitis. We analyze current research on the HIF pathway, elucidating its impact on bone repair and regeneration, while unraveling the involved cellular and molecular mechanisms. Furthermore, we briefly discuss the potential application of targeted interventions aimed at HIF in the field of bone tissue regeneration engineering. This review expands our biological understanding of the intricate relationship between the HIF gene and bone angiogenesis in periodontitis and offers valuable insights for the development of innovative therapies to expedite bone repair and regeneration.
Collapse
Affiliation(s)
- Chao Shan
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - YuNing Xia
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - Zeyu Wu
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - Jin Zhao
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China; Xinjiang Uygur Autonomous Region Institute of Stomatology, Ürümqi, China.
| |
Collapse
|
5
|
Hypoxia in Skin Cancer: Molecular Basis and Clinical Implications. Int J Mol Sci 2023; 24:ijms24054430. [PMID: 36901857 PMCID: PMC10003002 DOI: 10.3390/ijms24054430] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Skin cancer is one of the most prevalent cancers in the Caucasian population. In the United States, it is estimated that at least one in five people will develop skin cancer in their lifetime, leading to significant morbidity and a healthcare burden. Skin cancer mainly arises from cells in the epidermal layer of the skin, where oxygen is scarce. There are three main types of skin cancer: malignant melanoma, basal cell carcinoma, and squamous cell carcinoma. Accumulating evidence has revealed a critical role for hypoxia in the development and progression of these dermatologic malignancies. In this review, we discuss the role of hypoxia in treating and reconstructing skin cancers. We will summarize the molecular basis of hypoxia signaling pathways in relation to the major genetic variations of skin cancer.
Collapse
|
6
|
Hu Z, Cao Y, Galan EA, Hao L, Zhao H, Tang J, Sang G, Wang H, Xu B, Ma S. Vascularized Tumor Spheroid-on-a-Chip Model Verifies Synergistic Vasoprotective and Chemotherapeutic Effects. ACS Biomater Sci Eng 2022; 8:1215-1225. [PMID: 35167260 DOI: 10.1021/acsbiomaterials.1c01099] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prolyl hydroxylases (PHD) inhibitors have been observed to improve drug distribution in mice tumors via blood vessel normalization, increasing the effectiveness of chemotherapy. These effects are yet to be demonstrated in human cell models. Tumor spheroids are three-dimensional cell clusters that have demonstrated great potential in drug evaluation for personalized medicine. Here, we used a perfusable vascularized tumor spheroid-on-a-chip to simulate the tumor microenvironment in vivo and demonstrated that the PHD inhibitor dimethylallyl glycine prevents the degradation of normal blood vessels while enhancing the efficacy of the anticancer drugs paclitaxel and cisplatin in human esophageal carcinoma (Eca-109) spheroids. Our results point to the potential of this model to evaluate anticancer drugs under more physiologically relevant conditions.
Collapse
Affiliation(s)
- Zhiwei Hu
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| | - Yuanxiong Cao
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| | - Edgar A Galan
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| | - Liang Hao
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Haoran Zhao
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| | - Jiyuan Tang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| | - Gan Sang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| | - Hanqi Wang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| | - Bing Xu
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China.,Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China.,Shenzhen Bay Laboratory, Shenzhen 518107, China
| |
Collapse
|
7
|
Wen X, Peng Y, Gao M, Zhu Y, Zhu Y, Yu F, Zhou T, Shao J, Feng L, Ma X. Endothelial Transient Receptor Potential Canonical Channel Regulates Angiogenesis and Promotes Recovery After Myocardial Infarction. J Am Heart Assoc 2022; 11:e023678. [PMID: 35253458 PMCID: PMC9075314 DOI: 10.1161/jaha.121.023678] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background
Transient receptor potential canonical (TRPC) channels play a role in angiogenesis. However, the involvement of TRPC1 in myocardial infarction (MI) remains unclear. The present study was aimed at investigating whether TRPC1 can improve the recovery of cardiac function via prompting angiogenesis following MI.
Methods and Results
In vitro, coronary artery endothelial cells from floxed TRPC1 mice and endothelial cell‐specific TRPC1 channel knockout mice were cultured to access EC angiogenesis. Both EC tube formation and migration were significantly suppressed in mouse coronary artery endothelial cells from endothelial cell‐specific TRPC1 channel knockout mice. In vivo, coronary artery endothelial cells from floxed TRPC1 and endothelial cell‐specific TRPC1 channel knockout mice were subjected to MI, then echocardiography, triphenyltetrazolium chloride staining and immunofluorescence were performed to assess cardiac repair on day 28. Endothelial cell‐specific TRPC1 channel knockout mice had higher ejection fraction change, larger myocardial infarct size, and reduced capillary density in the infarct area compared with coronary artery endothelial cells from floxed TRPC1 mice. Furthermore, we found underlying regulation by HIF‐1α (hypoxic inducible factor‐1α) and MEK‐ERK (mitogen‐activated protein kinase/extracellular signal‐regulated kinase) that could be the mechanism for the angiogenetic action of TRPC1. Significantly, treatment with dimethyloxaloylglycine, an activator of HIF‐1α, induced cardiac improvement via the HIF‐1α‐TRPC1‐MEK/ERK pathway in MI mice.
Conclusions
Our study demonstrated TRPC1 improves cardiac function after MI by increasing angiogenesis via the upstream regulator HIF‐1α and downstream MEK/ERK, and dimethyloxaloylglycine treatment has protective effect on MI through the HIF‐1α‐TRPC1‐MEK/ERK pathway.
Collapse
Affiliation(s)
- Xin Wen
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Yidi Peng
- School of Pharmaceutical Sciences Jiangnan University Wuxi China
| | - Mengru Gao
- School of Pharmaceutical Sciences Jiangnan University Wuxi China
| | - Yuzhong Zhu
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Yifei Zhu
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Fan Yu
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Tingting Zhou
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Jing Shao
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Lei Feng
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Xin Ma
- Wuxi School of Medicine Jiangnan University Wuxi China
- School of Pharmaceutical Sciences Jiangnan University Wuxi China
| |
Collapse
|
8
|
Hutami IR, Izawa T, Khurel-Ochir T, Sakamaki T, Iwasa A, Tanaka E. Macrophage Motility in Wound Healing Is Regulated by HIF-1α via S1P Signaling. Int J Mol Sci 2021; 22:ijms22168992. [PMID: 34445695 PMCID: PMC8396560 DOI: 10.3390/ijms22168992] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 12/20/2022] Open
Abstract
Accumulating evidence indicates that the molecular pathways mediating wound healing induce cell migration and localization of cytokines to sites of injury. Macrophages are immune cells that sense and actively respond to disturbances in tissue homeostasis by initiating, and subsequently resolving, inflammation. Hypoxic conditions generated at a wound site also strongly recruit macrophages and affect their function. Hypoxia inducible factor (HIF)-1α is a transcription factor that contributes to both glycolysis and the induction of inflammatory genes, while also being critical for macrophage activation. For the latter, HIF-1α regulates sphingosine 1-phosphate (S1P) to affect the migration, activation, differentiation, and polarization of macrophages. Recently, S1P and HIF-1α have received much attention, and various studies have been performed to investigate their roles in initiating and resolving inflammation via macrophages. It is hypothesized that the HIF-1α/S1P/S1P receptor axis is an important determinant of macrophage function under inflammatory conditions and during disease pathogenesis. Therefore, in this review, biological regulation of monocytes/macrophages in response to circulating HIF-1α is summarized, including signaling by S1P/S1P receptors, which have essential roles in wound healing.
Collapse
Affiliation(s)
- Islamy Rahma Hutami
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan; (I.R.H.); (T.K.-O.); (T.S.); (A.I.); (E.T.)
- Department of Orthodontics, Faculty of Dentistry, Sultan Agung Islamic University, Semarang 50112, Indonesia
| | - Takashi Izawa
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan; (I.R.H.); (T.K.-O.); (T.S.); (A.I.); (E.T.)
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Correspondence: ; Tel.: +81-86-235-6691; Fax: +81-88-235-6694
| | - Tsendsuren Khurel-Ochir
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan; (I.R.H.); (T.K.-O.); (T.S.); (A.I.); (E.T.)
| | - Takuma Sakamaki
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan; (I.R.H.); (T.K.-O.); (T.S.); (A.I.); (E.T.)
| | - Akihiko Iwasa
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan; (I.R.H.); (T.K.-O.); (T.S.); (A.I.); (E.T.)
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan; (I.R.H.); (T.K.-O.); (T.S.); (A.I.); (E.T.)
| |
Collapse
|
9
|
Chang KH, Shoureshi P, Lay F, Sebastian R, Alikhassy Habibabady Z, Born LJ, Marti GP, Meltzer SJ, Abraham JM, Harmon JW. Preconditioning of surgical pedicle flaps with DNA plasmid expressing hypoxia-inducible factor-1α (HIF-1α) promotes tissue viability. Gene Ther 2021; 28:319-328. [PMID: 33024315 DOI: 10.1038/s41434-020-00199-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 09/05/2020] [Accepted: 09/24/2020] [Indexed: 01/29/2023]
Abstract
Ischemic necrosis of surgical flaps after reconstruction is a major clinical problem. Hypoxia-inducible factor-1α (HIF-1α) is considered the master regulator of the adaptive response to hypoxia. Among its many properties, it regulates the expression of genes encoding angiogenic growth factors, which have a short half-life in vivo. To achieve a continuous application of the therapeutic, we utilized DNA plasmid delivery. Transcription of the DNA plasmid confirmed by qRT-PCR showed significantly increased mRNA for HIF-1α in the transfected tissue compared to saline control tissue. Rats were preconditioned by injecting with either HIF-1α DNA plasmid or saline intradermally in the designated flap region on each flank. Seven days after preconditioning, each rat had two isolated pedicle flaps raised with a sterile silicone sheet implanted between the skin flap and muscle layer. The flaps preconditioned with HIF-1α DNA plasmid had significantly less necrotic area. Angiogenesis measured by CD31 staining showed a significant increase in the number of vessels per high powered field in the HIF-1α group (p < 0.05). Our findings offer a potential therapeutic strategy for significantly promoting the viability of surgical pedicle flaps by ischemic preconditioning with HIF-1α DNA plasmid.
Collapse
Affiliation(s)
- Kai-Hua Chang
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Pouria Shoureshi
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frank Lay
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raul Sebastian
- Department of Surgery, George Washington University School of Medicine and Health Science, Washington, DC, USA
| | | | - Louis J Born
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Guy P Marti
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Clinique Saint Jean l'Ermitage, Melun, France
| | - Stephen J Meltzer
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John M Abraham
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John W Harmon
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
10
|
Hutami IR, Izawa T, Khurel-Ochir T, Sakamaki T, Iwasa A, Tomita S, Tanaka E. HIF-1α controls palatal wound healing by regulating macrophage motility via S1P/S1P 1 signaling axis. Oral Dis 2021; 28:1157-1169. [PMID: 33759275 DOI: 10.1111/odi.13856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/01/2021] [Accepted: 03/16/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To investigate the role of hypoxia-inducible factor 1α (HIF-1α) signaling, the expression profile of M1 and M2 macrophages, and the role of the sphingosine 1-phosphate (S1P)/S1P receptor system in palatal wound healing of heterozygous HIF-1α-deficient (HIF-1α HET) mice. MATERIALS AND METHODS HIF-1α HET and wild-type (WT) littermates underwent palatal tissue excision at the mid-hard palate. Histological analysis, immunostaining, real-time PCR, Western blotting (WB), and cellular migration assays were performed to analyze wound closure and macrophage infiltration. RESULTS DMOG pretreatment showed an acceleration of palatal wound closure in WT mice. In contrast, the delayed palatal wound closure was observed in HIF-1α HET mice with diminished production of Col1a1, MCP-1, and MIP-1α, compared with WT mice. Decreased infiltration of M1 macrophage (F4/80+ TNF-α+ , F4/80+ iNOS+ ) and M2 macrophage (F4/80+ Arginase-1+ , F4/80+ CD163+ ) was observed. The numbers of F4/80+ S1P1 + macrophages of HIF-1α HET wounded tissues were significantly lower compared with WT tissues. S1P treatment of bone marrow macrophages (BMMs) significantly upregulated expression of S1P1 in WT mice compared with HIF-1α HET. Phosphorylation of MAPK rapidly decreased in BMMs of HIF-1α HET mice than in BMMs of WT mice by S1P stimulation. Moreover, S1P enhanced HIF-1α expression via S1P1 receptors to affect macrophage migration. CONCLUSIONS HIF-1α deficiency aggravates M1 and M2 macrophage infiltration and controls macrophage motility via S1P/S1P1 signaling. These results suggest that HIF-1α signaling may contribute to the regulation of palatal wound healing.
Collapse
Affiliation(s)
- Islamy Rahma Hutami
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.,Department of Orthodontics, Faculty of Dentistry, Sultan Agung Islamic University, Semarang, Jawa Tengah, Indonesia
| | - Takashi Izawa
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.,Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tsendsuren Khurel-Ochir
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takuma Sakamaki
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Akihiko Iwasa
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Shuhei Tomita
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
11
|
Rafique M, Wei T, Sun Q, Midgley AC, Huang Z, Wang T, Shafiq M, Zhi D, Si J, Yan H, Kong D, Wang K. The effect of hypoxia-mimicking responses on improving the regeneration of artificial vascular grafts. Biomaterials 2021; 271:120746. [PMID: 33725586 DOI: 10.1016/j.biomaterials.2021.120746] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/16/2021] [Accepted: 02/28/2021] [Indexed: 12/12/2022]
Abstract
Cellular transition to hypoxia following tissue injury, has been shown to improve angiogenesis and regeneration in multiple tissues. To take advantage of this, many hypoxia-mimicking scaffolds have been prepared, yet the oxygen access state of implanted artificial small-diameter vascular grafts (SDVGs) has not been investigated. Therefore, the oxygen access state of electrospun PCL grafts implanted into rat abdominal arteries was assessed. The regions proximal to the lumen and abluminal surfaces of the graft walls were normoxic and only the interior of the graft walls was hypoxic. In light of this differential oxygen access state of the implanted grafts and the critical role of vascular regeneration on SDVG implantation success, we investigated whether modification of SDVGs with HIF-1α stabilizer dimethyloxalylglycine (DMOG) could achieve hypoxia-mimicking responses resulting in improving vascular regeneration throughout the entirety of the graft wall. Therefore, DMOG-loaded PCL grafts were fabricated by electrospinning, to support the sustained release of DMOG over two weeks. In vitro experiments indicated that DMOG-loaded PCL mats had significant biological advantages, including: promotion of human umbilical vein endothelial cells (HUVECs) proliferation, migration and production of pro-angiogenic factors; and the stimulation of M2 macrophage polarization, which in-turn promoted macrophage regulation of HUVECs migration and smooth muscle cells (SMCs) contractile phenotype. These beneficial effects were downstream of HIF-1α stabilization in HUVECs and macrophages in normoxic conditions. Our results indicated that DMOG-loaded PCL grafts improved endothelialization, contractile SMCs regeneration, vascularization and modulated the inflammatory reaction of grafts in abdominal artery replacement models, thus promoting vascular regeneration.
Collapse
Affiliation(s)
- Muhammad Rafique
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Tingting Wei
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qiqi Sun
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Adam C Midgley
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ziqi Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ting Wang
- Tianjin Key Laboratory of Urban Transport Emission Research, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Muhammad Shafiq
- Department of Biotechnology, Faculty of Life Sciences, University of Central Punjab, Lahore, 54000, Pakistan
| | - Dengke Zhi
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jianghua Si
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Hongyu Yan
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Kai Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
12
|
Imanishi M, Yamakawa Y, Fukushima K, Ikuto R, Maegawa A, Izawa-Ishizawa Y, Horinouchi Y, Kondo M, Kishuku M, Goda M, Zamami Y, Takechi K, Chuma M, Ikeda Y, Tsuchiya K, Fujino H, Tsuneyama K, Ishizawa K. Fibroblast-specific ERK5 deficiency changes tumor vasculature and exacerbates tumor progression in a mouse model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:1239-1250. [PMID: 32307577 DOI: 10.1007/s00210-020-01859-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/20/2020] [Indexed: 01/12/2023]
Abstract
The roles of cancer-associated fibroblasts (CAFs) have been studied in the tumor progression, and CAFs are expected to become the new targets for cancer pharmacotherapies. CAFs contribute to tumor cell survival and proliferation, tumor angiogenesis, immune suppression, tumor inflammation, tumor cell invasion and metastasis, and extracellular matrix remodeling. However, detailed mechanisms of how CAFs function in the living system remain unclear. CAFs include α-smooth muscle actin, expressing activated fibroblasts similar to myofibroblasts, and are highly capable of producing collagen. Several reports have demonstrated the contributions of extracellular-signal-regulated kinase 5 (ERK5) in fibroblasts to the fibrotic processes; however, the roles of CAF-derived ERK5 remain unclear. To investigate the roles of CAF-derived ERK5 in the tumor progression, we created mice lacking the ERK5 gene specifically in fibroblasts. Colon-26 mouse colon cancer cells were implanted into the mice subcutaneously, and the histological analyses of the tumor tissue were performed after 2 weeks. Immunofluorescence analyses showed that recipient-derived fibroblasts existed within the tumor tissue. The present study demonstrated that fibroblast-specific ERK5 deficiency exacerbated tumor progression and it was accompanied with thicker tumor vessel formation and the increase in the number of activated fibroblasts. We combined the results of The Cancer Genome Atlas (TCGA) database analysis with our animal studies, and indicated that regulating ERK5 activity in CAFs or CAF invasion into the tumor tissue can be important strategies for the development of new targets in cancer pharmacotherapies.
Collapse
Affiliation(s)
- Masaki Imanishi
- Department of Pharmacy, Tokushima University Hospital, 2-50-1, Kuramoto-cho, Tokushima, 770-8503, Japan.
| | - Yusuke Yamakawa
- Department of Pharmacy, Tokushima University Hospital, 2-50-1, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Keijo Fukushima
- Department of Pharmacology for Life Sciences, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Raiki Ikuto
- Department of Clinical Pharmacology and Therapeutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Akiko Maegawa
- Department of Clinical Pharmacology and Therapeutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | | | - Yuya Horinouchi
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Masateru Kondo
- Department of Pharmacy, Tokushima University Hospital, 2-50-1, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Masatoshi Kishuku
- Department of Pharmacy, Tokushima University Hospital, 2-50-1, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Mitsuhiro Goda
- Department of Pharmacy, Tokushima University Hospital, 2-50-1, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Yoshito Zamami
- Department of Pharmacy, Tokushima University Hospital, 2-50-1, Kuramoto-cho, Tokushima, 770-8503, Japan
- Department of Clinical Pharmacology and Therapeutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kenshi Takechi
- Clinical Trial Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Masayuki Chuma
- Clinical Trial Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hiromichi Fujino
- Department of Pharmacology for Life Sciences, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Pharmacy, Tokushima University Hospital, 2-50-1, Kuramoto-cho, Tokushima, 770-8503, Japan
- AWA Support Center, Tokushima University, Tokushima, Japan
| |
Collapse
|
13
|
Matsunaga S, Tomita S. [The effect of PHD inhibitor on tumor microenvironment and tumor immune response]. Nihon Yakurigaku Zasshi 2020; 155:35-39. [PMID: 31902845 DOI: 10.1254/fpj.19119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Tumor tissue environment is generally exposed to low oxygen, nutrition depletion and high interstitial pressure condition. These circumstances are caused by vascular hyper-permeability, irregular vascularization and immature vessels. The blood vessel is important tissue structures to deliver oxygen, nutrition and so on. An abnormal blood vessel formation is a common feature of tumor tissue that were characterized by hyper-permeability, irregular vascularization, immature vessels and intravasation. Therefore, tumor tissue is exposed to low oxygen nutrition depletion and low pH due to hypoperfusion and elevated interstitial pressure. These environments are one of the reasons for chemo- and radio-resistance. Previously, we reported that prolyl hydroxylase (PHD) inhibitor induced tumor blood vessel normalization and improved tumor microenvironment in tumor mouse model. However, effects of PHD inhibitor on tumor progression is controversial. Enhanced hypoxia inducible factors (HIFs) signaling in cancer cells act to promote cancer proliferation and metastases. On the other hand, increasing of HIFs signaling in immune cells may lead to activate inflammation and elicit anti-tumor effect. We describe our study how PHD inhibitor improved tumor microenvironment and focused on tumor infiltrate immune cells were phenotypic alteration after PHD inhibitor treatment in mouse model. Our results implied that PHD inhibitor was possibly beneficial for anti-cancer therapy.
Collapse
Affiliation(s)
- Shinji Matsunaga
- Department of Pharmacology, Osaka City University Graduate School of Medicine
| | - Shuhei Tomita
- Department of Pharmacology, Osaka City University Graduate School of Medicine
| |
Collapse
|
14
|
Nishide S, Matsunaga S, Shiota M, Yamaguchi T, Kitajima S, Maekawa Y, Takeda N, Tomura M, Uchida J, Miura K, Nakatani T, Tomita S. Controlling the Phenotype of Tumor-Infiltrating Macrophages via the PHD-HIF Axis Inhibits Tumor Growth in a Mouse Model. iScience 2019; 19:940-954. [PMID: 31518902 PMCID: PMC6742914 DOI: 10.1016/j.isci.2019.08.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/01/2019] [Accepted: 08/20/2019] [Indexed: 01/02/2023] Open
Abstract
The tumor microenvironment (TME) polarizes tumor-infiltrating macrophages toward tumor support. Macrophage-abundant tumors are highly malignant and are the cause of poor prognosis and therapeutic resistance. In this study, we show that the prolyl hydroxylase (PHD) inhibitor FG-4592 (FG) inhibits tumor growth of macrophage-abundant tumors and prolongs mouse survival. FG not only normalizes tumor vessels and improves tumor oxygenation but also directly affects macrophages and activates phagocytosis through the PHD-hypoxia-inducible factor (HIF) axis. Remarkably, FG can promote phagocytic ability of the Ly6Clo subset of tumor-infiltrating macrophages, leading to tumor growth inhibition. Moreover, Ly6Cneg macrophages contributed to blood vessel normalization. Using a malignant tumor mouse model, we characterized macrophage function and subsets. Altogether, our findings suggest that the PHD inhibitor can promote the anti-tumor potential of macrophages to improve cancer therapy. PHD inhibitor treatment inhibits tumor growth and prolongs survival time of mice Regulating the PHD-HIF pathway can alter the tumor-infiltrating macrophage phenotype PHD inhibitor activates the tumor phagocytic ability of Ly6Clo macrophages
Collapse
Affiliation(s)
- Shunji Nishide
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan; Department of Urology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Shinji Matsunaga
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Masayuki Shiota
- Division of Research Support Platform, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Takehiro Yamaguchi
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Shojiro Kitajima
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Yoichi Maekawa
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Domain of Integrated Life Systems, Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University, Gifu 501-1193, Japan
| | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka 584-8540, Japan
| | - Junji Uchida
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Katsuyuki Miura
- Department of Applied Pharmacology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Tatsuya Nakatani
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Shuhei Tomita
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan.
| |
Collapse
|
15
|
Perioperative Treatment with a Prolyl Hydroxylase Inhibitor Reduces Necrosis in a Rat Ischemic Skin Flap Model. Plast Reconstr Surg 2019; 143:769e-779e. [DOI: 10.1097/prs.0000000000005441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Discussion: Perioperative Treatment with a Prolyl Hydroxylase Inhibitor Reduces Necrosis in a Rat Ischemic Skin Flap Model. Plast Reconstr Surg 2019; 143:780e-781e. [PMID: 30921128 DOI: 10.1097/prs.0000000000005442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Zhu M, Liu X, Wang Y, Chen L, Wang L, Qin X, Xu J, Li L, Tu Y, Zhou T, Sang A, Song E. YAP via interacting with STAT3 regulates VEGF-induced angiogenesis in human retinal microvascular endothelial cells. Exp Cell Res 2018; 373:155-163. [DOI: 10.1016/j.yexcr.2018.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/10/2018] [Accepted: 10/12/2018] [Indexed: 12/29/2022]
|
18
|
Koyama S, Matsunaga S, Imanishi M, Maekawa Y, Kitano H, Takeuchi H, Tomita S. Tumour blood vessel normalisation by prolyl hydroxylase inhibitor repaired sensitivity to chemotherapy in a tumour mouse model. Sci Rep 2017; 7:45621. [PMID: 28361934 PMCID: PMC5374523 DOI: 10.1038/srep45621] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 02/17/2017] [Indexed: 12/28/2022] Open
Abstract
Blood vessels are important tissue structures that deliver oxygen and nutrition. In tumour tissue, abnormal blood vessels, which are hyperpermeable and immature, are often formed; these tissues also have irregular vascularisation and intravasation. This situation leads to hypoperfusion in tumour tissue along with low oxygen and nutrition depletion; this is also called the tumour microenvironment and is characterised by hypoxia, depleted nutrition, low pH and high interstitial pressure. This environment induces resistance to anticancer drugs, which causes an increase in anticancer drug doses, leading to increased side effects. We hypothesised that normalised tumour blood vessels would improve tumour tissue perfusion, resupply nutrition and re-oxygenate the tumour tissue. Chemotherapy would then be more effective and cause a decrease in anticancer drug doses. Here we report a neovascularisation-inducing drug that improved tumour vascular abnormalities, such as low blood flow, blood leakage and abnormal vessel structure. These results could lead to not only an increased chemo-sensitivity and tissue-drug distribution but also an up-regulated efficiency for cancer chemotherapy. This suggests that tumour blood vessel normalisation therapy accompanied by angiogenesis may be a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Satoshi Koyama
- Division of Molecular Pharmacology, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Japan.,Division of Otolaryngology, Head and Neck Surgery, Department of Sensory and Motor Organs, Tottori University Faculty of Medicine, Japan
| | - Shinji Matsunaga
- Division of Molecular Pharmacology, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Japan.,Department of Pharmacology, Osaka City University Graduate School of Medicine, Japan
| | - Masaki Imanishi
- Division of Molecular Pharmacology, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Japan
| | - Yoichi Maekawa
- Department of Parasitology Gifu University Graduate School of Medicine, Japan.,Domain of Integrated Life Systems, Gifu Center for Highly Advanced Integration of Nanosciences and Life Sciences (G-CHAIN), Gifu University, Gifu, Japan
| | - Hiroya Kitano
- Division of Otolaryngology, Head and Neck Surgery, Department of Sensory and Motor Organs, Tottori University Faculty of Medicine, Japan
| | - Hiromi Takeuchi
- Division of Otolaryngology, Head and Neck Surgery, Department of Sensory and Motor Organs, Tottori University Faculty of Medicine, Japan
| | - Shuhei Tomita
- Division of Molecular Pharmacology, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Japan.,Department of Pharmacology, Osaka City University Graduate School of Medicine, Japan
| |
Collapse
|
19
|
Fukunaga Y, Izawa-Ishizawa Y, Horinouchi Y, Sairyo E, Ikeda Y, Ishizawa K, Tsuchiya K, Abe Y, Hashimoto I, Tamaki T. Topical application of nitrosonifedipine, a novel radical scavenger, ameliorates ischemic skin flap necrosis in a mouse model. Wound Repair Regen 2017; 25:217-223. [PMID: 28090711 DOI: 10.1111/wrr.12510] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/10/2016] [Accepted: 01/10/2017] [Indexed: 12/23/2022]
Abstract
Ischemic skin flap necrosis can occur in random pattern flaps. An excess amount of reactive oxygen species is generated and causes necrosis in the ischemic tissue. Nitrosonifedipine (NO-NIF) has been demonstrated to possess potent radical scavenging ability. However, there has been no study on the effects of NO-NIF on ischemic skin flap necrosis. Therefore, they evaluated the potential of NO-NIF in ameliorating ischemic skin flap necrosis in a mouse model. A random pattern skin flap (1.0 × 3.0 cm) was elevated on the dorsum of C57BL/6 mice. NO-NIF was administered by topical injection immediately after surgery and every 24 hours thereafter. Flap survival was evaluated on postoperative day 7. Tissue samples from the skin flaps were harvested on postoperative days 1 and 3 to analyze oxidative stress, apoptosis and endothelial dysfunction. The viable area of the flap in the NO-NIF group was significantly increased (78.30 ± 7.041%) compared with that of the control group (47.77 ± 6.549%, p < 0.01). NO-NIF reduced oxidative stress, apoptosis and endothelial dysfunction, which were evidenced by the decrease of malondialdehyde, p22phox protein expression, number of apoptotic cells, phosphorylated p38 MAPK protein expression, and vascular cell adhesion molecule-1 protein expression while endothelial nitric oxide synthase protein expression was increased. In conclusion, they demonstrated that NO-NIF ameliorated ischemic skin flap necrosis by reducing oxidative stress, apoptosis, and endothelial dysfunction. NO-NIF is considered to be a candidate for the treatment of ischemic flap necrosis.
Collapse
Affiliation(s)
- Yutaka Fukunaga
- Department of Plastic and Reconstructive Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Yuki Izawa-Ishizawa
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Yuya Horinouchi
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Eriko Sairyo
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Keisuke Ishizawa
- Department of Pharmacy, Tokushima University Hospital.,Department of Clinical Pharmacy, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yoshiro Abe
- Department of Plastic and Reconstructive Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Ichiro Hashimoto
- Department of Plastic and Reconstructive Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Toshiaki Tamaki
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School
| |
Collapse
|
20
|
MiRNA-Sequence Indicates That Mesenchymal Stem Cells and Exosomes Have Similar Mechanism to Enhance Cardiac Repair. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4150705. [PMID: 28203568 PMCID: PMC5292186 DOI: 10.1155/2017/4150705] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/21/2016] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) repair infarcted heart through paracrine mechanism. We sought to compare the effectiveness of MSCs and MSC-derived exosomes (MSC-Exo) in repairing infarcted hearts and to identify how MSC-Exo mediated cardiac repair is regulated. In a rat myocardial infarction model, we found that MSC-Exo inhibited cardiac fibrosis, inflammation, and improved cardiac function. The beneficial effects of MSC-Exo were significantly superior compared to that of MSCs. To explore the potential mechanisms underlying MSC-Exo's effects, we performed several in vitro experiments and miRNA-sequence analysis. MSC-Exo stimulated cardiomyocyte H9C2 cell proliferation, inhibited apoptosis induced by H2O2, and inhibited TGF-β induced transformation of fibroblast cell into myofibroblast. Importantly, novel miRNA sequencing results indicated that MSC-Exo and MSCs have similar miRNA expression profile, which could be one of the reasons that MSC-Exo can replace MSCs for cardiac repair. In addition, the expression of several miRNAs from MSC-Exo was significantly different from that of MSCs, which may explain why MSC-Exo has better therapeutic effect than MSCs. In conclusion, this study demonstrates that MSC-Exo could be used alone to promote cardiac repair and are superior to MSCs in repairing injured myocardium.
Collapse
|
21
|
Mayorga M, Kiedrowski M, Shamhart P, Forudi F, Weber K, Chilian WM, Penn MS, Dong F. Early upregulation of myocardial CXCR4 expression is critical for dimethyloxalylglycine-induced cardiac improvement in acute myocardial infarction. Am J Physiol Heart Circ Physiol 2015; 310:H20-8. [PMID: 26519029 DOI: 10.1152/ajpheart.00449.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/04/2015] [Indexed: 12/23/2022]
Abstract
The stromal cell-derived factor-1 (SDF-1):CXCR4 is important in myocardial repair. In this study we tested the hypothesis that early upregulation of cardiomyocyte CXCR4 (CM-CXCR4) at a time of high myocardial SDF-1 expression could be a strategy to engage the SDF-1:CXCR4 axis and improve cardiac repair. The effects of the hypoxia inducible factor (HIF) hydroxylase inhibitor dimethyloxalylglycine (DMOG) on CXCR4 expression was tested on H9c2 cells. In mice a myocardial infarction (MI) was produced in CM-CXCR4 null and wild-type controls. Mice were randomized to receive injection of DMOG (DMOG group) or saline (Saline group) into the border zone after MI. Protein and mRNA expression of CM-CXCR4 were quantified. Echocardiography was used to assess cardiac function. During hypoxia, DMOG treatment increased CXCR4 expression of H9c2 cells by 29 and 42% at 15 and 24 h, respectively. In vivo DMOG treatment increased CM-CXCR4 expression at 15 h post-MI in control mice but not in CM-CXCR4 null mice. DMOG resulted in increased ejection fraction in control mice but not in CM-CXCR4 null mice 21 days after MI. Consistent with greater cardiomyocyte survival with DMOG treatment, we observed a significant increase in cardiac myosin-positive area within the infarct zone after DMOG treatment in control mice, but no increase in CM-CXCR4 null mice. Inhibition of cardiomyocyte death in MI through the stabilization of HIF-1α requires downstream CM-CXCR4 expression. These data suggest that engagement of the SDF-1:CXCR4 axis through the early upregulation of CM-CXCR4 is a strategy for improving cardiac repair after MI.
Collapse
Affiliation(s)
- Mari Mayorga
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, and
| | - Matthew Kiedrowski
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, and
| | - Patricia Shamhart
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, and
| | - Farhad Forudi
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, and
| | - Kristal Weber
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, and
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, and
| | - Marc S Penn
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, and Summa Cardiovascular Institute, Summa Health System, Akron, Ohio
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, and
| |
Collapse
|
22
|
Esfahani M, Karimi F, Afshar S, Niknazar S, Sohrabi S, Najafi R. Prolyl hydroxylase inhibitors act as agents to enhance the efficiency of cell therapy. Expert Opin Biol Ther 2015; 15:1739-55. [PMID: 26325448 DOI: 10.1517/14712598.2015.1084281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION In stem cell-based therapy as a subtype of regenerative medicine, stem cells can be used to replace or repair injured tissue and cells in order to treat disease. Stem cells have the ability to integrate into injured areas and produce new cells via processes of proliferation and differentiation. Several studies have demonstrated that hypoxia increases self-renewal, proliferation and post-homing differentiation of stem cells through the regulation of hypoxia-inducible factor-1 (HIF-1)-mediated gene expression. Thus, pharmacological interventions including prolyl hydroxylase (PHD) inhibitors are considered as promising solutions for stem cell-based therapy. PHD inhibitors stabilize the HIF-1 and activate its pathway through preventing proteasomal degradation of HIF-1. AREAS COVERED This review focuses on the role of hypoxia, HIF-1 and especially PHD inhibitors on cell therapy. PHD structure and function are discussed as well as their inhibitors. In addition, we have investigated several preclinical studies in which PHD inhibitors improved the efficiency of cell-based therapies. EXPERT OPINION The data reviewed here suggest that PHD inhibitors are effective operators in improving stem cell therapy. However, because of some limitations, these compounds should be properly examined before clinical application.
Collapse
Affiliation(s)
- Maryam Esfahani
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Fatemeh Karimi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Saeid Afshar
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Somayeh Niknazar
- b 2 Shahid Beheshti University of Medical Science, Hearing Disorders Research Center , Tehran, the Islamic Republic of Iran
| | - Sareh Sohrabi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Rezvan Najafi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| |
Collapse
|
23
|
Harnoss JM, Strowitzki MJ, Radhakrishnan P, Platzer LK, Harnoss JC, Hank T, Cai J, Ulrich A, Schneider M. Therapeutic inhibition of prolyl hydroxylase domain-containing enzymes in surgery: putative applications and challenges. HYPOXIA 2015; 3:1-14. [PMID: 27774478 PMCID: PMC5045068 DOI: 10.2147/hp.s60872] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Oxygen is essential for metazoans to generate energy. Upon oxygen deprivation adaptive and protective pathways are induced, mediated by hypoxia-inducible factors (HIFs) and prolyl hydroxylase domain-containing enzymes (PHDs). Both play a pivotal role in various conditions associated with prolonged ischemia and inflammation, and are promising targets for therapeutic intervention. This review focuses on aspects of therapeutic PHD modulation in surgically relevant disease conditions such as hepatic and intestinal disorders, wound healing, innate immune responses, and tumorigenesis, and discusses the therapeutic potential and challenges of PHD inhibition in surgical patients.
Collapse
Affiliation(s)
- Jonathan Michael Harnoss
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Moritz Johannes Strowitzki
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Lisa Katharina Platzer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Julian Camill Harnoss
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Hank
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jun Cai
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
24
|
|
25
|
Peng J, Lai ZG, Fang ZL, Xing S, Hui K, Hao C, Jin Q, Qi Z, Shen WJ, Dong QN, Bing ZH, Fu DL. Dimethyloxalylglycine prevents bone loss in ovariectomized C57BL/6J mice through enhanced angiogenesis and osteogenesis. PLoS One 2014; 9:e112744. [PMID: 25394221 PMCID: PMC4231053 DOI: 10.1371/journal.pone.0112744] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/12/2014] [Indexed: 12/16/2022] Open
Abstract
Hypoxia-inducible factor 1-α (HIF-1α) plays a critical role in angiogenesis-osteogenesis coupling during bone development and bone regeneration. Previous studies have shown that 17β-estradiol activates the HIF-1α signaling pathway and that mice with conditional activation of the HIF-1α signaling pathway in osteoblasts are protected from ovariectomy (OVX)-induced bone loss. In addition, it has been shown that hypoxia facilitates the osteogenic differentiation of mesenchymal stem cells (MSCs) and modulates Wnt/β-catenin signaling. Therefore, we hypothesized that activation of the HIF-1α signaling pathway by hypoxia-mimicking agents would prevent bone loss due to estrogen deficiency. In this study, we confirmed the effect of dimethyloxalylglycine (DMOG), a hypoxia-mimicking agent, on the HIF-1α signaling pathway and investigated the effect of DMOG on MSC osteogenic differentiation and the Wnt/β-catenin signaling pathway. We then investigated the effect of DMOG treatment on OVX-induced bone loss. Female C57BL/6J mice were divided into sham, OVX, OVX+L-DMOG (5 mg/kg/day), and OVX+H-DMOG (20 mg/kg/day) groups. At sacrifice, static and dynamic bone histomorphometry were performed with micro computed tomography (micro-CT) and undecalcified sections, respectively. Bone strength was assessed with the three-point bending test, and femur vessels were reconstructed and analyzed by micro-CT. Serum vascular endothelial growth factor (VEGF), osteocalcin, and C-terminal telopeptides of collagen type(CTX) were measured by ELISA. Tartrate-resistant acid phosphatase staining was used to assess osteoclast formation. Alterations in the HIF-1α and Wnt/β-catenin signaling pathways in the bone were detected by western blot. Our results showed that DMOG activated the HIF-1α signaling pathway, which further activated the Wnt/β-catenin signaling pathway and enhanced MSC osteogenic differentiation. The micro-CT results showed that DMOG treatment improved trabecular bone density and restored the bone microarchitecture and blood vessels in OVX mice. Bone strength was also partly restored in DMOG-treated OVX mice. Dynamic bone histomorphometric analysis of the femur metaphysic revealed that DMOG increased the mineralizing surface, mineral apposition rate, and bone formation rate. The serum levels of VEGF and osteocalcin were higher in DMOG-treated OVX mice. However, there were no significant differences in serum CTX or in the number of tartrate-resistant acid phosphatase-stained cells between DMOG-treated OVX mice and OVX mice. Western blot results showed that DMOG administration partly rescued the decrease in HIF-1α and β-catenin expression following ovariectomy. Collectively, these results indicate that DMOG prevents bone loss due to ovariectomy in C57BL/6J mice by enhancing angiogenesis and osteogenesis, which are associated with activated HIF-1α and Wnt/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Jia Peng
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Zuo Gui Lai
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
- Department of Orthopaedics, Qian Fo Shan Hospital, Shang Dong University, Ji Nan, China
| | - Zhang Lian Fang
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shen Xing
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Kang Hui
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Hao
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jin
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Zhou Qi
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Wang Jin Shen
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Nian Dong
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Zhou Han Bing
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Deng Lian Fu
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
26
|
Liu XB, Wang JA, Ji XY, Yu SP, Wei L. Preconditioning of bone marrow mesenchymal stem cells by prolyl hydroxylase inhibition enhances cell survival and angiogenesis in vitro and after transplantation into the ischemic heart of rats. Stem Cell Res Ther 2014; 5:111. [PMID: 25257482 PMCID: PMC4535299 DOI: 10.1186/scrt499] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 07/16/2014] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION Poor cell survival and limited functional benefits have restricted the efficacy of bone marrow mesenchymal stem cells (BMSCs) in the treatment of myocardial infarction. We showed recently that hypoxia preconditioning of BMSCs and neural progenitor cells before transplantation can enhance the survival and therapeutic properties of these cells in the ischemic brain and heart. The present investigation explores a novel strategy of preconditioning BMSCs using the Hypoxia-inducible factor 1α (HIF-α) prolyl hydroxylase inhibitor dimethyloxalylglycine (DMOG) to enhance their survival and therapeutic efficacy after transplantation into infarcted myocardium. METHODS BMSCs from green fluorescent protein transgenic rats were cultured with or without 1 mM DMOG for 24 hours in complete culture medium before transplantation. Survival and angiogenic factors were evaluated in vitro by trypan blue staining, Western blotting, and tube formation test. In an ischemic heart model of rats, BMSCs with and without DMOG preconditioning were intramyocardially transplanted into the peri-infarct region 30 minutes after permanent myocardial ischemia. Cell death was measured 24 hours after engraftment. Heart function, angiogenesis and infarct size were measured 4 weeks later. RESULTS In DMOG preconditioned BMSCs (DMOG-BMSCs), the expression of survival and angiogenic factors including HIF-1α, vascular endothelial growth factor, glucose transporter 1 and phospho-Akt were significantly increased. In comparison with control cells, DMOG-BMSCs showed higher viability and enhanced angiogenesis in both in vitro and in vivo assays. Transplantation of DMOG-BMSCs reduced heart infarct size and promoted functional benefits of the cell therapy. CONCLUSIONS We suggest that DMOG preconditioning enhances the survival capability of BMSCs and paracrine effects with increased differentiation potential. Prolyl hydroxylase inhibition is an effective and feasible strategy to enhance therapeutic efficacy and efficiency of BMSC transplantation therapy after heart ischemia.
Collapse
|
27
|
Kuchler U, Keibl C, Fügl A, Schwarze UY, Tangl S, Agis H, Gruber R. Dimethyloxalylglycine lyophilized onto bone substitutes increase vessel area in rat calvarial defects. Clin Oral Implants Res 2014; 26:485-91. [DOI: 10.1111/clr.12474] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Ulrike Kuchler
- Department of Oral Surgery; Medical University of Vienna; Vienna Austria
- Department of Oral Surgery and Stomatology; University of Berne; Berne Switzerland
| | - Claudia Keibl
- Austrian Cluster for Tissue Regeneration; Vienna Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology; Vienna Austria
| | - Alexander Fügl
- Department of Oral Surgery; Medical University of Vienna; Vienna Austria
- Austrian Cluster for Tissue Regeneration; Vienna Austria
| | - Uwe Y. Schwarze
- Department of Oral Surgery; Medical University of Vienna; Vienna Austria
- Austrian Cluster for Tissue Regeneration; Vienna Austria
- Karl Donath Laboratory for Hard Tissue and Biomaterial Research; Department of Oral Surgery; Medical University of Vienna; Vienna Austria
| | - Stefan Tangl
- Department of Oral Surgery; Medical University of Vienna; Vienna Austria
- Austrian Cluster for Tissue Regeneration; Vienna Austria
- Karl Donath Laboratory for Hard Tissue and Biomaterial Research; Department of Oral Surgery; Medical University of Vienna; Vienna Austria
| | - Hermann Agis
- Austrian Cluster for Tissue Regeneration; Vienna Austria
- Department of Conservative Dentistry and Periodontology; Medical University of Vienna; Vienna Austria
| | - Reinhard Gruber
- Department of Oral Surgery; Medical University of Vienna; Vienna Austria
- Department of Oral Surgery and Stomatology; University of Berne; Berne Switzerland
- Laboratory of Oral Cell Biology; School of Dental Medicine; University of Berne; Berne Switzerland
| |
Collapse
|
28
|
Selvaraju V, Parinandi NL, Adluri RS, Goldman JW, Hussain N, Sanchez JA, Maulik N. Molecular mechanisms of action and therapeutic uses of pharmacological inhibitors of HIF-prolyl 4-hydroxylases for treatment of ischemic diseases. Antioxid Redox Signal 2014; 20:2631-65. [PMID: 23992027 PMCID: PMC4026215 DOI: 10.1089/ars.2013.5186] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 08/06/2013] [Accepted: 09/01/2013] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE In this review, we have discussed the efficacy and effect of small molecules that act as prolyl hydroxylase domain inhibitors (PHDIs). The use of these compounds causes upregulation of the pro-angiogenic factors and hypoxia inducible factor-1α and -2α (HIF-1α and HIF-2α) to enhance angiogenic, glycolytic, erythropoietic, and anti-apoptotic pathways in the treatment of various ischemic diseases responsible for significant morbidity and mortality in humans. RECENT ADVANCES Sprouting of new blood vessels from the existing vasculature and surgical intervention, such as coronary bypass and stent insertion, have been shown to be effective in attenuating ischemia. However, the initial reentry of oxygen leads to the formation of reactive oxygen species that cause oxidative stress and result in ischemia/reperfusion (IR) injury. This apparent "oxygen paradox" must be resolved to combat IR injury. During hypoxia, decreased activity of PHDs initiates the accumulation and activation of HIF-1α, wherein the modulation of both PHD and HIF-1α appears as promising therapeutic targets for the pharmacological treatment of ischemic diseases. CRITICAL ISSUES Research on PHDs and HIFs has shown that these molecules can serve as therapeutic targets for ischemic diseases by modulating glycolysis, erythropoiesis, apoptosis, and angiogenesis. Efforts are underway to identify and synthesize safer small-molecule inhibitors of PHDs that can be administered in vivo as therapy against ischemic diseases. FUTURE DIRECTIONS This review presents a comprehensive and current account of the existing small-molecule PHDIs and their use in the treatment of ischemic diseases with a focus on the molecular mechanisms of therapeutic action in animal models.
Collapse
Affiliation(s)
- Vaithinathan Selvaraju
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Narasimham L. Parinandi
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio
| | - Ram Sudheer Adluri
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Joshua W. Goldman
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Naveed Hussain
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, Connecticut
- Division of Neonatal Medicine, Connecticut Children's Medical Center, Hartford, Connecticut
| | - Juan A. Sanchez
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Nilanjana Maulik
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
29
|
Overexpressed HIF-2α in Endothelial Cells Promotes Vascularization and Improves Random Pattern Skin Flap Survival. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2014; 2:e132. [PMID: 25289325 PMCID: PMC4174206 DOI: 10.1097/gox.0000000000000083] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 04/26/2014] [Indexed: 02/05/2023]
Abstract
BACKGROUND The local skin flap procedure is very useful for reconstruction. However, flap necrosis caused by circulatory failure can occur at its distal portion. Hypoxia-inducible factors (HIFs) in endothelial cells (ECs) help to maintain ECs and promote vascularization, and HIF-2α is abundantly expressed in ECs. However, the mechanisms of action of HIF-2α in ECs are not yet fully understood. The aim of this study was to evaluate the in vivo effects of overexpression of HIF-2α in ECs on skin flap survival. METHODS A random pattern skin flap (1.0 × 3.0 cm) was elevated on the dorsum of transgenic mice (Tg mice) with EC-specific HIF-2α conditional overexpression and wild-type littermate control mice (n = 6). Flap survival was evaluated on postoperative day 7. Tissue samples from the skin flaps were harvested and analyzed using Western blotting, quantitative reverse transcriptase-polymerase chain reaction, and immunohistochemistry. RESULTS The HIF-2α mRNA and protein levels were significantly increased in the Tg mice when compared with control mice. Tg mice had significantly increased skin flap survival areas (72.0% ± 2.7%) when compared with wild-type mice (45.7% ± 1.1%). Moreover, histological examination revealed an increase in the subcutaneous blood vessel counts in the Tg mice. CONCLUSIONS Specific overexpression of HIF-2α in ECs promoted vascularization and enhanced skin flap survival in vivo in a mouse model.
Collapse
|
30
|
Hadjipanayi E, Schilling AF. Hypoxia-based strategies for angiogenic induction: the dawn of a new era for ischemia therapy and tissue regeneration. Organogenesis 2013; 9:261-72. [PMID: 23974216 PMCID: PMC3903695 DOI: 10.4161/org.25970] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Therapeutic angiogenesis promises to aid the healing and regeneration of tissues suffering from a compromised vascular supply. Ischaemia therapy has so far primarily focused on delivering isolated angiogenic growth factors. The limited success of these strategies in clinical trials, however, is increasingly forcing researchers to recognize the difficulties associated with trying to mimic the angiogenic process, due to its natural complexity. Instead, a new school of thought is gradually emerging, focusing on how to induce angiogenesis at its onset, by utilizing hypoxia, the primary angiogenic stimulus in physiological, as well pathological states. This shift in therapeutic approach is underlined by the realization of the importance of depressed HIF-1 α-mediated gene programming in non-healing ischemic tissues, which could explain their apparent habituation to chronic hypoxic stress and the limited capacity to generate adaptive angiogenesis. Hypoxia-based strategies, then effectively aim to override the habituated angiogenic cellular response, re-start the regenerative process and drive it to completion. Here we make a distinction between those strategies that utilize hypoxia in vitro as a preconditioning tool to optimize the angiogenic potential of tissue/cells before transplantation, vs. strategies that aim to induce hypoxia-induced signaling in vivo, directly, through pharmacological means or gene transfer. We then discuss possible future directions for the field, as it moves into the phase of clinical trials.
Collapse
Affiliation(s)
- Ektoras Hadjipanayi
- Experimental Plastic Surgery; Clinic for Plastic and Hand Surgery; Klinikum Rechts der Isar; Technische Universität München; Munich, Germany; Department of Plastic, Reconstructive, Hand and Burn Surgery; Bogenhausen Hospital; Munich, Germany
| | - Arndt F Schilling
- Experimental Plastic Surgery; Clinic for Plastic and Hand Surgery; Klinikum Rechts der Isar; Technische Universität München; Munich, Germany; Center for Applied New Technologies in Engineering for Regenerative Medicine (Canter); Munich, Germany
| |
Collapse
|
31
|
Myllyharju J. Prolyl 4-hydroxylases, master regulators of the hypoxia response. Acta Physiol (Oxf) 2013; 208:148-65. [PMID: 23489300 DOI: 10.1111/apha.12096] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 11/07/2012] [Accepted: 03/08/2013] [Indexed: 12/13/2022]
Abstract
A decrease in oxygenation is a life-threatening situation for most organisms. An evolutionarily conserved efficient and rapid hypoxia response mechanism activated by a hypoxia-inducible transcription factor (HIF) is present in animals ranging from the simplest multicellular phylum Placozoa to humans. In humans, HIF induces the expression of more than 100 genes that are required to increase oxygen delivery and to reduce oxygen consumption. As its name indicates HIF is found at protein level only in hypoxic cells, whereas in normoxia, it is degraded by the proteasome pathway. Prolyl 4-hydroxylases, enzymes that require oxygen in their reaction, are the cellular oxygen sensors regulating the stability of HIF. In normoxia, 4-hydroxyproline residues formed in the α-subunit of HIF by these enzymes lead to its ubiquitination by the von Hippel-Lindau E3 ubiquitin ligase and immediate destruction in proteasomes thus preventing the formation of a functional HIF αβ dimer. Prolyl 4-hydroxylation is inhibited in hypoxia, facilitating the formation of the HIF dimer and activation of its target genes, such as those for erythropoietin and vascular endothelial growth factor. This review starts with a summary of the molecular and catalytic properties and individual functions of the four HIF prolyl 4-hydroxylase isoenzymes. Induction of the hypoxia response via inhibition of the HIF prolyl 4-hydroxylases may provide a novel therapeutic target in the treatment of hypoxia-associated diseases. The current status of studies aiming at such therapeutic approaches is introduced in the final part of this review.
Collapse
Affiliation(s)
- J. Myllyharju
- Oulu Center for Cell-Matrix Research; Biocenter Oulu and Department of Medical Biochemistry and Molecular Biology; University of Oulu; Oulu; Finland
| |
Collapse
|
32
|
Ding H, Gao YS, Hu C, Wang Y, Wang CG, Yin JM, Sun Y, Zhang CQ. HIF-1α transgenic bone marrow cells can promote tissue repair in cases of corticosteroid-induced osteonecrosis of the femoral head in rabbits. PLoS One 2013; 8:e63628. [PMID: 23675495 PMCID: PMC3652809 DOI: 10.1371/journal.pone.0063628] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 04/04/2013] [Indexed: 02/01/2023] Open
Abstract
Although corticosteroid-induced osteonecrosis of the femoral head (ONFH) is common, the treatment for it remains limited and largely ineffective. We examined whether implantation of hypoxia inducible factor-1α (HIF-1α) transgenic bone marrow cells (BMCs) can promote the repair of the necrotic area of corticosteroid-induced ONFH. In this study, we confirmed that HIF-1α gene transfection could enhance mRNA expression of osteogenic genes in BMCs in vitro. Alkaline phosphatase activity assay and alizarin red-S staining indicated HIF-1α transgenic BMCs had enhanced osteogenic differentiation capacity in vitro. Furthermore, enzyme linked immunosorbent assay (ELISA) for VEGF revealed HIF-1α transgenic BMCs secreted more VEGF as compared to normal BMCs. An experimental rabbit model of early-stage corticosteroid-induced ONFH was established and used for an evaluation of cytotherapy. Transplantation of HIF-1α transgenic BMCs dramatically improved the bone regeneration of the necrotic area of the femoral head. The number and volume of blood vessel were significantly increased in the necrotic area of the femoral head compared to the control groups. These results support HIF-1α transgenic BMCs have enhanced osteogenic and angiogenic activity in vitro and in vivo. Transplantation of HIF-1α transgenic BMCs can potentially promote the repair of the necrotic area of corticosteroid-induced ONFH.
Collapse
Affiliation(s)
- Hao Ding
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - You-Shui Gao
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Hu
- Shanghai Key Laboratory of Regulatory Biology, School of life Sciences, East China Normal University, Shanghai, China
| | - Yang Wang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chuan-Gui Wang
- Shanghai Key Laboratory of Regulatory Biology, School of life Sciences, East China Normal University, Shanghai, China
| | - Ji-Min Yin
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Sun
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chang-Qing Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- * E-mail:
| |
Collapse
|