1
|
Van den Hoven P, S Weller F, Van De Bent M, Goncalves LN, Ruig M, D Van Den Berg S, Ooms S, Mieog J, Ea Van De Bogt K, Van Schaik J, Schepers A, Vahrmeijer AL, Hamming JF, Van Der Vorst JR. Near-infrared fluorescence imaging with indocyanine green for quantification of changes in tissue perfusion following revascularization. Vascular 2021; 30:867-873. [PMID: 34320878 DOI: 10.1177/17085381211032826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Current diagnostic modalities for patients with peripheral artery disease (PAD) mainly focus on the macrovascular level. For assessment of tissue perfusion, near-infrared (NIR) fluorescence imaging using indocyanine green (ICG) seems promising. In this prospective cohort study, ICG NIR fluorescence imaging was performed pre- and post-revascularization to assess changes in foot perfusion. METHODS ICG NIR fluorescence imaging was performed in 36 patients with PAD pre- and post-intervention. After intravenous bolus injection of 0.1 mg/kg ICG, the camera registered the NIR fluorescence intensity over time on the dorsum of the feet for 15 min using the Quest Spectrum Platform®. Time-intensity curves were plotted for three regions of interest (ROI): (1) the dorsum of the foot, (2) the forefoot, and (3) the hallux. Time-intensity curves were normalized for maximum fluorescence intensity. Extracted parameters were the maximum slope, area under the curve (AUC) for the ingress, and the AUC for the egress. The non-treated contralateral leg was used as a control group. RESULTS Successful revascularization was performed in 32 patients. There was a significant increase for the maximum slope and AUC egress in all three ROIs. The most significant difference was seen for the maximum slope in ROI 3 (3.7%/s to 6.6%/s, p < 0.001). In the control group, no significant differences were seen for the maximum slope and AUC egress in all ROIs. CONCLUSIONS This study shows the potential of ICG NIR fluorescence imaging in assessing the effect of revascularization procedures on foot perfusion. Future studies should focus on the use of this technique in predicting favorable outcome of revascularization procedures.
Collapse
Affiliation(s)
- Pim Van den Hoven
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Floris S Weller
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Merel Van De Bent
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Lauren N Goncalves
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Melissa Ruig
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Simen D Van Den Berg
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Sophie Ooms
- Department of Vascular Surgery, Haga Ziekenhuis, The Hague, The Netherlands
| | - Jsd Mieog
- Department of Surgical Oncology, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Koen Ea Van De Bogt
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Van Schaik
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Abbey Schepers
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Alex L Vahrmeijer
- Department of Surgical Oncology, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap F Hamming
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Joost R Van Der Vorst
- Department of Vascular Surgery, 4501Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Park J, Lee YK, Park IK, Hwang SR. Current Limitations and Recent Progress in Nanomedicine for Clinically Available Photodynamic Therapy. Biomedicines 2021; 9:85. [PMID: 33467201 PMCID: PMC7830249 DOI: 10.3390/biomedicines9010085] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Photodynamic therapy (PDT) using oxygen, light, and photosensitizers has been receiving great attention, because it has potential for making up for the weakness of the existing therapies such as surgery, radiation therapy, and chemotherapy. It has been mainly used to treat cancer, and clinical tests for second-generation photosensitizers with improved physicochemical properties, pharmacokinetic profiles, or singlet oxygen quantum yield have been conducted. Progress is also being made in cancer theranostics by using fluorescent signals generated by photosensitizers. In order to obtain the effective cytotoxic effects on the target cells and prevent off-target side effects, photosensitizers need to be localized to the target tissue. The use of nanocarriers combined with photosensitizers can enhance accumulation of photosensitizers in the tumor site, owing to preferential extravasation of nanoparticles into the tumor vasculature by the enhanced permeability and retention effect. Self-assembly of amphiphilic polymers provide good loading efficiency and sustained release of hydrophobic photosensitizers. In addition, prodrug nanomedicines for PDT can be activated by stimuli in the tumor site. In this review, we introduce current limitations and recent progress in nanomedicine for PDT and discuss the expected future direction of research.
Collapse
Affiliation(s)
- Jooho Park
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea;
| | - Yong-Kyu Lee
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea;
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Seung Rim Hwang
- College of Pharmacy, Chosun University, Gwangju 61452, Korea
| |
Collapse
|
3
|
van den Hoven P, Ooms S, van Manen L, van der Bogt KEA, van Schaik J, Hamming JF, Vahrmeijer AL, van der Vorst JR, Mieog JSD. A systematic review of the use of near-infrared fluorescence imaging in patients with peripheral artery disease. J Vasc Surg 2020; 70:286-297.e1. [PMID: 31230648 DOI: 10.1016/j.jvs.2018.11.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 11/19/2018] [Indexed: 01/18/2023]
Abstract
OBJECTIVE In the diagnosis of peripheral artery disease (PAD), the ankle-brachial index plays an important role. However, results of the ankle-brachial index are unreliable in patients with severe media sclerosis. Near-infrared (NIR) fluorescence imaging using indocyanine green (ICG) can provide information about tissue perfusion and has already been studied in oncologic, reconstructive, and cardiac surgery. For patients with PAD, this technique might give insight into skin perfusion and thereby guide treatment. We performed a systematic review of the literature on the use of NIR fluorescence imaging in patients with PAD. METHODS PubMed, MEDLINE, Embase, and Cochrane were searched for articles and abstracts on the application of NIR fluorescence imaging using ICG as fluorescent dye in patients with PAD. Our search strategy combined the terms "fluorescence," "ICG," or synonyms and "peripheral artery disease" or synonyms. The extracted data included fluorescence parameters and test characteristics for diagnosis of PAD. RESULTS Twenty-three articles were found eligible for this review using 18 different parameters for evaluation of the fluorescence signal intensity. NIR fluorescence imaging was used for four main indications: diagnosis, quality control in revascularization, guidance in amputation surgery, and visualization of vascular structures. For the diagnosis of PAD, NIR fluorescence imaging yields a sensitivity ranging from 67% to 100% and a specificity varying between 72% and 100%. Significant increases in multiple fluorescence parameters were found in comparing patients before and after revascularization. CONCLUSIONS NIR fluorescence imaging can be used for several indications in patients with PAD. NIR fluorescence imaging seems promising in diagnosis of PAD and guidance of surgeons in treatment, especially in patients in whom current diagnostic methods are not applicable. Further standardization is needed to reliably use this modality in patients with PAD.
Collapse
Affiliation(s)
- Pim van den Hoven
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Sophie Ooms
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Labrinus van Manen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jan van Schaik
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap F Hamming
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
4
|
Wang Y, Zhan Y, Harris LM, Khan S, Xia J. A portable three-dimensional photoacoustic tomography system for imaging of chronic foot ulcers. Quant Imaging Med Surg 2019; 9:799-806. [PMID: 31281775 DOI: 10.21037/qims.2019.05.02] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background Chronic leg ulcers affect approximately 6.5 million Americans and the disorder is associated with a range of serious complications. Since many chronic ulcers have underlying vascular insufficiency, accurate assessment of tissue perfusion is critical to treatment planning and post-surgical monitoring. However, existing clinical tests fail to meet this need in practice due to their low sensitivity or accuracy. Methods In this paper, we introduce a portable photoacoustic tomography (PAT) system for wound assessment. Since hemoglobin serves as the major endogenous contrast at near-infrared wavelengths, PAT provides label-free, three-dimensional (3D) imaging of hemoglobin distribution, which is closely related to blood perfusion. The proposed system consists of a 128-element linear transducer array, a data acquisition (DAQ) system, and a pulsed Nd:YAG laser source, all mounted on a portable cart for easy clinical testing. Results We validated our system through both phantom and human imaging studies. The phantom imaging results indicate that the system's spatial resolution ranges from 0.5 mm along the axial direction to 1.3 mm along the elevational direction. The healthy volunteer result shows clear foot vasculature, indicating good perfusion. The preliminary patient imaging results agree very well with the clinical test, demonstrating that PAT has a high potential for assessing the circulation around the wound. Conclusions We believe that our technique will be a valuable tool for assessing tissue perfusion and guiding wound treatment in vascular clinics.
Collapse
Affiliation(s)
- Yuehang Wang
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Ye Zhan
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Linda M Harris
- Department of Surgery, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Sikandar Khan
- Department of Surgery, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Jun Xia
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York, USA
| |
Collapse
|
5
|
Rho SY, Kim SH, Kang CM, Lee WJ. Is ICG-enhanced image able to help predicting pancreatic fistula in laparoscopic pancreaticoduodenectomy? MINIM INVASIV THER 2018; 28:29-32. [DOI: 10.1080/13645706.2018.1479271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Seoung Yoon Rho
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Pancreatobiliary Cancer Clinic, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
| | - Sung Hyun Kim
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Pancreatobiliary Cancer Clinic, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
| | - Chang Moo Kang
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Pancreatobiliary Cancer Clinic, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
| | - Woo Jung Lee
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Pancreatobiliary Cancer Clinic, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
| |
Collapse
|
6
|
Miller J, Wang ST, Orukari I, Prior J, Sudlow G, Su X, Liang K, Tang R, Hillman EM, Weilbaecher KN, Culver JP, Berezin MY, Achilefu S. Perfusion-based fluorescence imaging method delineates diverse organs and identifies multifocal tumors using generic near-infrared molecular probes. JOURNAL OF BIOPHOTONICS 2018; 11:e201700232. [PMID: 29206348 PMCID: PMC5903995 DOI: 10.1002/jbio.201700232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/03/2017] [Accepted: 12/03/2017] [Indexed: 06/07/2023]
Abstract
Rapid detection of multifocal cancer without the use of complex imaging schemes will improve treatment outcomes. In this study, dynamic fluorescence imaging was used to harness differences in the perfusion kinetics of near-infrared (NIR) fluorescent dyes to visualize structural characteristics of different tissues. Using the hydrophobic nontumor-selective NIR dye cypate, and the hydrophilic dye LS288, a high tumor-to-background contrast was achieved, allowing the delineation of diverse tissue types while maintaining short imaging times. By clustering tissue types with similar perfusion properties, the dynamic fluorescence imaging method identified secondary tumor locations when only the primary tumor position was known, with a respective sensitivity and specificity of 0.97 and 0.75 for cypate, and 0.85 and 0.81 for LS288. Histological analysis suggests that the vasculature in the connective tissue that directly surrounds the tumor was a major factor for tumor identification through perfusion imaging. Although the hydrophobic dye showed higher specificity than the hydrophilic probe, use of other dyes with different physical and biological properties could further improve the accuracy of the dynamic imaging platform to identify multifocal tumors for potential use in real-time intraoperative procedures.
Collapse
Affiliation(s)
- Jessica Miller
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
- Biomedical Engineering, Washington University in St. Louis, 1 Brookings Dr., St. Louis, Missouri 63130, United States
| | - Steven T. Wang
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
| | - Inema Orukari
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
- Biomedical Engineering, Washington University in St. Louis, 1 Brookings Dr., St. Louis, Missouri 63130, United States
| | - Julie Prior
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
| | - Gail Sudlow
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
| | - Xinming Su
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Kexian Liang
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
| | - Rui Tang
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
| | - Elizabeth M.C. Hillman
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Ave., New York, NY 10027, United States
| | - Katherine N. Weilbaecher
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Joseph P. Culver
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
- Biomedical Engineering, Washington University in St. Louis, 1 Brookings Dr., St. Louis, Missouri 63130, United States
| | - Mikhail Y. Berezin
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
- Department of Chemistry, Washington University, St. Louis, Missouri 63132, United States
| | - Samuel Achilefu
- Optical Radiology Lab, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, Missouri 63110, United States
- Biomedical Engineering, Washington University in St. Louis, 1 Brookings Dr., St. Louis, Missouri 63130, United States
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
7
|
An Y, Kang Y, Lee J, Ahn C, Kwon K, Choi C. Blood flow characteristics of diabetic patients with complications detected by optical measurement. Biomed Eng Online 2018; 17:25. [PMID: 29466988 PMCID: PMC5822764 DOI: 10.1186/s12938-018-0457-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/16/2018] [Indexed: 12/12/2022] Open
Abstract
Background Diabetes mellitus (DM) is one of the most common diseases worldwide. Uncontrolled and prolonged hyperglycemia can cause diabetic complications, which reduce the quality of life of patients. Diabetic complications are common in DM patients. Because it is impossible to completely recover from diabetic complications, it is important for early detection. In this study, we suggest a novel method of determining blood flow characteristics based on fluorescence image analysis with indocyanine green and report that diabetic complications have unique blood flow characteristics. Methods We analyzed time-series fluorescence images obtained from controls, DM patients, and DM patients with complications. The images were segmented into the digits and the dorsum of the feet and hands, and each part has been considered as arterial and capillary flow. We compared the blood flow parameters in each region among the three groups. Results The DM patients with complications showed similar blood flow parameters to the controls, except the area under the curve and the maximum intensity, which indicate the blood flow volume. These parameters were significantly decreased in DM patients with complications. Although some blood flow parameters in the feet of DM patients with complications were close to normal blood flow, the vascular response of the macrovessels and microvessels to stimulation of the hands was significantly reduced, which indicates less reactivity in DM patients with complications. Conclusions Our results suggest that DM patients, and DM patients with complications, have unique peripheral blood flow characteristics.
Collapse
Affiliation(s)
- Yuri An
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Yujung Kang
- R&D Center, Vieworks Co., Anyang-si, Gyeonggi-do, Republic of Korea
| | - Jungsul Lee
- Cellex Life Sciences, Inc, Daejeon, Republic of Korea
| | - Chulwoo Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kihwan Kwon
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Chulhee Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea. .,Cellex Life Sciences, Inc, Daejeon, Republic of Korea.
| |
Collapse
|
8
|
Jagtap J, Sharma G, Parchur AK, Gogineni V, Bergom C, White S, Flister MJ, Joshi A. Methods for detecting host genetic modifiers of tumor vascular function using dynamic near-infrared fluorescence imaging. BIOMEDICAL OPTICS EXPRESS 2018; 9:543-556. [PMID: 29552392 PMCID: PMC5854057 DOI: 10.1364/boe.9.000543] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/07/2017] [Accepted: 01/03/2018] [Indexed: 05/06/2023]
Abstract
Vascular supply is a critical component of the tumor microenvironment (TME) and is essential for tumor growth and metastasis, yet the endogenous genetic modifiers that impact vascular function in the TME are largely unknown. To identify the host TME modifiers of tumor vascular function, we combined a novel genetic mapping strategy [Consomic Xenograft Model] with near-infrared (NIR) fluorescence imaging and multiparametric analysis of pharmacokinetic modeling. To detect vascular flow, an intensified cooled camera based dynamic NIR imaging system with 785 nm laser diode based excitation was used to image the whole-body fluorescence emission of intravenously injected indocyanine green dye. Principal component analysis was used to extract the spatial segmentation information for the lungs, liver, and tumor regions-of-interest. Vascular function was then quantified by pK modeling of the imaging data, which revealed significantly altered tissue perfusion and vascular permeability that were caused by host genetic modifiers in the TME. Collectively, these data demonstrate that NIR fluorescent imaging can be used as a non-invasive means for characterizing host TME modifiers of vascular function that have been linked with tumor risk, progression, and response to therapy.
Collapse
Affiliation(s)
- Jaidip Jagtap
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Gayatri Sharma
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Abdul K. Parchur
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Carmen Bergom
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sarah White
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael J. Flister
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amit Joshi
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
9
|
Cai W, Li Y, Liu F, Luo J. Quantitative evaluation of graded hindlimb ischemia based on pharmacokinetic modelling and hemodynamic analysis of indocyanine green. Physiol Meas 2018; 39:015009. [PMID: 29231185 DOI: 10.1088/1361-6579/aaa108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Accurate evaluation of the degree of hindlimb ischemia is challenging but essential for the diagnosis and treatment of peripheral vascular insufficiency. The aim of the study is to apply a multiparametric method for the quantitative estimation of mouse models with different degrees of hindlimb ischemia based on a dynamic fluorescence imaging-based strategy. APPROACH An adjustable hydraulic occluder was placed around the thigh root of one hindlimb to induce six different degrees of hindlimb ischemia. Five parameters were extracted to quantitatively evaluate the degree of ischemia, including perfusion rate (PR) and perfusion vascular density (PVD) from a mathematical model of indocyanine green (ICG) pharmacokinetics, rising time (T rise), blood flow index (BFI) and mean fluorescence intensity (MFI) from time-series analysis of ICG hemodynamics. MAIN RESULTS The results showed that the normalized PR and BFI decreased while the normalized T rise increased progressively with the degree of ischemia. The normalized PVD and MFI first increased and then decreased with the degree of ischemia. High correlation was observed between the degree of ischemia and the arterial oxygen saturation which was measured by an oximeter. SIGNIFICANCE The results of this work demonstrated that PR, BFI and T rise can be used for the quantitative and comprehensive evaluation of graded hindlimb ischemia.
Collapse
Affiliation(s)
- Wenjuan Cai
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | | | | | | |
Collapse
|
10
|
Zahavi-Goldstein E, Blumenfeld M, Fuchs-Telem D, Pinzur L, Rubin S, Aberman Z, Sher N, Ofir R. Placenta-derived PLX-PAD mesenchymal-like stromal cells are efficacious in rescuing blood flow in hind limb ischemia mouse model by a dose- and site-dependent mechanism of action. Cytotherapy 2017; 19:1438-1446. [PMID: 29122516 DOI: 10.1016/j.jcyt.2017.09.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND In peripheral artery disease (PAD), blockage of the blood supply to the limbs, most frequently the legs, leads to impaired blood flow and tissue ischemia. Pluristem's PLX-PAD cells are placenta-derived mesenchymal stromal-like cells currently in clinical trials for the treatment of peripheral artery diseases. METHODS In this work, the hind limb ischemia (HLI) mouse model was utilized to study the efficacy and mechanism of action of PLX-PAD cells. ELISA assays were performed to characterize and quantitate PLX-PAD secretions in vitro. RESULTS PLX-PAD cells administered intramuscularly rescued blood flow to the lower limb after HLI induction in a dose-dependent manner. While rescue of blood flow was site-dependent, numerous administration regimes enabled rescue of blood flow, indicating a systemic effect mediated by PLX-PAD secretions. Live PLX-PAD cells were more efficacious than cell lysate in rescuing blood flow, indicating the importance of prolonged cytokine secretion for maximal blood flow recovery. In vitro studies showed a multifactorial secretion profile including numerous pro-angiogenic proteins; these are likely involved in the PLX-PAD mechanism of action. DISCUSSION Live PLX-PAD cells were efficacious in rescuing blood flow after the induction of HLI in the mouse model in a dose- and site-dependent manner. The fact that various administration routes of PLX-PAD rescued blood flow indicates that the mechanism of action likely involves one of systemic secretions which promote angiogenesis. Taken together, the data support the further clinical testing of PLX-PAD cells for PAD indications.
Collapse
|
11
|
Hwang Y, Yoon H, Choe K, Ahn J, Jung JH, Park JH, Kim P. In vivo cellular-level real-time pharmacokinetic imaging of free-form and liposomal indocyanine green in liver. BIOMEDICAL OPTICS EXPRESS 2017; 8:4706-4716. [PMID: 29082096 PMCID: PMC5654811 DOI: 10.1364/boe.8.004706] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 05/18/2023]
Abstract
Indocyanine green (ICG) is a near-infrared fluorophore approved for human use which has been widely used for various clinical applications. Despite the well-established clinical usage, our understanding about the microscopic in vivo pharmacokinetics of systemically administered ICG has been relatively limited. In this work, we successfully visualized real-time in vivo pharmacokinetic dynamics of the intravenously injected free-form and liposomal ICG in cellular resolution by utilizing a custom-built video-rate near infrared laser-scanning confocal microscopy system. Initial perfusion and clearance from blood stream, diffusion into perisinusoidal space, and subsequent absorption into hepatocyte were directly visualized in vivo. The quantification analysis utilizing the real-time image sequences revealed distinct dynamic in vivo pharmacokinetic behavior of free-form and liposomal ICG.
Collapse
Affiliation(s)
- Yoonha Hwang
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Hwanjun Yoon
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Kibaek Choe
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Jinhyo Ahn
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Jik Han Jung
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Ji-Ho Park
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Pilhan Kim
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| |
Collapse
|
12
|
Herold J, Nowak S, Kostin S, Daniel JM, Francke A, Subramaniam S, Braun-Dullaeus RC, Kanse SM. Factor VII activating protease (FSAP) influences vascular remodeling in the mouse hind limb ischemia model. Am J Transl Res 2017; 9:3084-3095. [PMID: 28670395 PMCID: PMC5489907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/09/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Investigations in factor VII activating protease (FSAP)-/- mice suggest a role for FSAP in stroke, thrombosis and neointima formation. Here, we analyzed the role of FSAP in vascular remodeling processes related to arteriogenesis and angiogenesis in the mouse hind limb ischemia model. METHODS AND RESULTS Femoral artery ligation was performed in mice and exogenous FSAP was injected locally to examine its effect on arteriogenesis in the adductor and angiogenesis in the gastrocnemius muscle over 21 days. Perfusion was decreased by FSAP, which was reflected in a lower arterial diameter and was associated with reduced monocyte infiltration in the adductor muscle. There was increased angiogenesis in the gastrocnemius muscle triggered indirectly by less blood supply to the lower limb. Comparison of wild-type (WT) and FSAP-/- mice showed that perfusion was not different between the genotypes but there were 2.5-fold more collateral arteries in the adductor muscle of FSAP-/- mice at day 21. This was associated with a higher infiltration of monocytes at day 3. Capillary density in the gastrocnemius muscle was not altered. Activity of the two major proteolytic pathways associated with vascular remodeling; matrix metalloprotease (MMP)-9 and urokinase-type plasminogen activator (uPA) was elevated in the gastrocnemius but not in the adductor muscle in FSAP-/- mice. CONCLUSIONS Arteriogenesis is enhanced, and this is associated with a higher infiltration of monocytes, in the absence of endogenous FSAP but angiogenesis is unchanged. Exogenous FSAP had the opposite effect on arteriogenesis indicating a possible therapeutic potential of modulating endogenous FSAP.
Collapse
Affiliation(s)
- Joerg Herold
- Department of Internal Medicine and Cardiology, University of MagdeburgGermany
| | - Steven Nowak
- Department of Internal Medicine and Cardiology, University of MagdeburgGermany
| | | | | | - Alexander Francke
- Department of Internal Medicine and Cardiology, University of MagdeburgGermany
| | | | | | | |
Collapse
|
13
|
Cai W, Guang H, Cai C, Luo J. Effects of temperature on multiparametric evaluation of hindlimb ischemia with dynamic fluorescence imaging. JOURNAL OF BIOPHOTONICS 2017; 10:811-820. [PMID: 27925417 DOI: 10.1002/jbio.201600235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/31/2016] [Accepted: 11/22/2016] [Indexed: 05/09/2023]
Abstract
Quantitative evaluation of hindlimb ischemia is essential for early diagnosis and therapy of peripheral arterial disease (PAD). Dynamic imaging using near-infrared (NIR) fluorophore indocyanine green (ICG) is a noninvasive and effective tool to monitor multiple vascular parameters including perfusion rate (PR), perfusion vascular density (PVD) and hemodynamics. It has been previously demonstrated that temperature changes could lead to significant variations of blood flow rate and vascular perfusion. In this paper, multiparametric evaluation of hindlimb ischemia was performed at different temperatures. Five different parameters were extracted from dynamic fluorescence imaging, including PR, PVD, rising time (Trise ), blood flow index (BFI) and mean fluorescence intensity (MFI). Temperatures varied from 15 °C to 40 °C were set on a mouse model of hindlimb ischemia. The aforementioned five parameters were obtained at each temperature. The results suggest that PVD, BFI and MFI could be effective indicators to distinguish ischemic tissues from normal tissues in mouse hindlimb at different temperatures. In contrast, PR is effective only when the temperature is higher than 25 °C, while Trise is effective only when the temperature is lower than 35 °C. The parameters of PVD, BFI and MFI could provide quantitative and comprehensive evaluation for PAD at different temperatures. (A) Bright-field image of the normal (left) and ischemic (right) hindlimbs. (B-D) Parametric images of perfusion vascular density, blood flow index and mean fluorescence intensity.
Collapse
Affiliation(s)
- Wenjuan Cai
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Huizhi Guang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Chuangjian Cai
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Jianwen Luo
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
- Center for Biomedical Imaging Research, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
14
|
Guang H, Cai C, Zuo S, Cai W, Zhang J, Luo J. Multiparametric evaluation of hindlimb ischemia using time-series indocyanine green fluorescence imaging. JOURNAL OF BIOPHOTONICS 2017; 10:456-464. [PMID: 27135903 DOI: 10.1002/jbio.201600029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/03/2016] [Accepted: 04/10/2016] [Indexed: 06/05/2023]
Abstract
Peripheral arterial disease (PAD) can further cause lower limb ischemia. Quantitative evaluation of the vascular perfusion in the ischemic limb contributes to diagnosis of PAD and preclinical development of new drug. In vivo time-series indocyanine green (ICG) fluorescence imaging can noninvasively monitor blood flow and has a deep tissue penetration. The perfusion rate estimated from the time-series ICG images is not enough for the evaluation of hindlimb ischemia. The information relevant to the vascular density is also important, because angiogenesis is an essential mechanism for post-ischemic recovery. In this paper, a multiparametric evaluation method is proposed for simultaneous estimation of multiple vascular perfusion parameters, including not only the perfusion rate but also the vascular perfusion density and the time-varying ICG concentration in veins. The target method is based on a mathematical model of ICG pharmacokinetics in the mouse hindlimb. The regression analysis performed on the time-series ICG images obtained from a dynamic reflectance fluorescence imaging system. The results demonstrate that the estimated multiple parameters are effective to quantitatively evaluate the vascular perfusion and distinguish hypo-perfused tissues from well-perfused tissues in the mouse hindlimb. The proposed multiparametric evaluation method could be useful for PAD diagnosis. The estimated perfusion rate and vascular perfusion density maps (left) and the time-varying ICG concentration in veins of the ankle region (right) of the normal and ischemic hindlimbs.
Collapse
Affiliation(s)
- Huizhi Guang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Chuangjian Cai
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Simin Zuo
- Department of Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, 52074, Germany
| | - Wenjuan Cai
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Jiulou Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Jianwen Luo
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
- Center for Biomedical Imaging Research, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
15
|
Venermo M, Settembre N, Albäck A, Vikatmaa P, Aho PS, Lepäntalo M, Inoue Y, Terasaki H. Pilot Assessment of the Repeatability of Indocyanine Green Fluorescence Imaging and Correlation with Traditional Foot Perfusion Assessments. Eur J Vasc Endovasc Surg 2016; 52:527-533. [PMID: 27486005 DOI: 10.1016/j.ejvs.2016.06.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/28/2016] [Indexed: 01/06/2023]
Abstract
BACKGROUND Ankle brachial index (ABI), toe pressures (TP), and transcutaneous oxygen pressure (TcPO2) are traditionally used in the assessment of critical limb ischemia (CLI). Indocyanine green (ICG) fluorescence imaging can be used to evaluate local circulation in the foot and to evaluate the severity of ischemia. This prospective study analyzed the suitability of a fluorescence imaging system (photodynamic eye [PDE]) in CLI. MATERIAL AND METHODS Forty-one patients with CLI were included. Of the patients, 66% had diabetes and there was an ischemic tissue lesion in 70% of the limbs. ABI, toe pressures, TcPO2 and ICG-fluorescence imaging (ICG-FI) were measured in each leg. To study the repeatability of the ICG-FI, each patient underwent the study twice. After the procedure, foot circulation was measured using a time-intensity curve, where T1/2 (the time needed to achieve half of the maximum fluorescence intensity) and PDE10 (increase of the intensity during the first 10 s) were determined. A time-intensity curve was plotted using the same areas as for the TcPO2 probes (n=123). RESULTS The mean ABI was 0.43, TP 21 mmHg, TcPO2 23 mmHg, T1/2 38 s, and PDE10 19 AU. Time-intensity curves were repeatable. In a Bland-Altman scatter plot, the 95% limits of agreement of PDE10 was 9.9 AU and the corresponding value of T1/2 was 14 s. Correlation between ABI and TP was significant (R=.73, p<.001), and it was weaker in diabetic patients (R=.47, p=.048) compared with non-diabetic patients (R=.89, p=.002). Correlations between ABI and TcPO2 and TP and TcPO2 were weak (R=.37, p=.05 and R=.43, p=.037, respectively). Correlation between TcPO2 and PDE10 was strong in diabetic patients (R=.70, p=.003). CONCLUSIONS According to this pilot study, ICG-FI with PDE can be used in the assessment of blood supply in the ischemic foot.
Collapse
Affiliation(s)
- M Venermo
- Helsinki University Central Hospital, Helsinki, Finland.
| | - N Settembre
- Helsinki University Central Hospital, Helsinki, Finland
| | - A Albäck
- Helsinki University Central Hospital, Helsinki, Finland
| | - P Vikatmaa
- Helsinki University Central Hospital, Helsinki, Finland
| | - P-S Aho
- Helsinki University Central Hospital, Helsinki, Finland
| | - M Lepäntalo
- Helsinki University Central Hospital, Helsinki, Finland
| | - Y Inoue
- Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - H Terasaki
- Helsinki University Central Hospital, Helsinki, Finland; Tokyo Medical and Dental University Hospital, Tokyo, Japan
| |
Collapse
|
16
|
Gan Q, Wang D, Ye J, Zhang Z, Wang X, Hu C, Shao P, Xu RX. Benchtop and Animal Validation of a Projective Imaging System for Potential Use in Intraoperative Surgical Guidance. PLoS One 2016; 11:e0157794. [PMID: 27391764 PMCID: PMC4938571 DOI: 10.1371/journal.pone.0157794] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 06/03/2016] [Indexed: 11/18/2022] Open
Abstract
We propose a projective navigation system for fluorescence imaging and image display in a natural mode of visual perception. The system consists of an excitation light source, a monochromatic charge coupled device (CCD) camera, a host computer, a projector, a proximity sensor and a Complementary metal-oxide-semiconductor (CMOS) camera. With perspective transformation and calibration, our surgical navigation system is able to achieve an overall imaging speed higher than 60 frames per second, with a latency of 330 ms, a spatial sensitivity better than 0.5 mm in both vertical and horizontal directions, and a projection bias less than 1 mm. The technical feasibility of image-guided surgery is demonstrated in both agar-agar gel phantoms and an ex vivo chicken breast model embedding Indocyanine Green (ICG). The biological utility of the system is demonstrated in vivo in a classic model of ICG hepatic metabolism. Our benchtop, ex vivo and in vivo experiments demonstrate the clinical potential for intraoperative delineation of disease margin and image-guided resection surgery.
Collapse
Affiliation(s)
- Qi Gan
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
| | - Dong Wang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Jian Ye
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
| | - Zeshu Zhang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
| | - Xinrui Wang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
| | - Chuanzhen Hu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
| | - Pengfei Shao
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
- * E-mail: (PS); (RXX)
| | - Ronald X. Xu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (PS); (RXX)
| |
Collapse
|
17
|
Seo J, An Y, Lee J, Ku T, Kang Y, Ahn C, Choi C. Principal component analysis of dynamic fluorescence images for diagnosis of diabetic vasculopathy. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:46003. [PMID: 27071414 DOI: 10.1117/1.jbo.21.4.046003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/22/2016] [Indexed: 05/09/2023]
Abstract
Indocyanine green (ICG) fluorescence imaging has been clinically used for noninvasive visualizations of vascular structures. We have previously developed a diagnostic system based on dynamic ICG fluorescence imaging for sensitive detection of vascular disorders. However, because high-dimensional raw data were used, the analysis of the ICG dynamics proved difficult. We used principal component analysis (PCA) in this study to extract important elements without significant loss of information. We examined ICG spatiotemporal profiles and identified critical features related to vascular disorders. PCA time courses of the first three components showed a distinct pattern in diabetic patients. Among the major components, the second principal component (PC2) represented arterial-like features. The explained variance of PC2 in diabetic patients was significantly lower than in normal controls. To visualize the spatial pattern of PCs, pixels were mapped with red, green, and blue channels. The PC2 score showed an inverse pattern between normal controls and diabetic patients. We propose that PC2 can be used as a representative bioimaging marker for the screening of vascular diseases. It may also be useful in simple extractions of arterial-like features.
Collapse
Affiliation(s)
- Jihye Seo
- Korea Advanced Institute of Science and Technology, Department of Bio and Brain Engineering, Daejeon 34141, Korea
| | - Yuri An
- Korea Advanced Institute of Science and Technology, Department of Bio and Brain Engineering, Daejeon 34141, KoreabKorea Advanced Institute of Science and Technology, KI for the BioCentury, Daejeon 34141, Korea
| | - Jungsul Lee
- Korea Advanced Institute of Science and Technology, Department of Bio and Brain Engineering, Daejeon 34141, KoreabKorea Advanced Institute of Science and Technology, KI for the BioCentury, Daejeon 34141, Korea
| | - Taeyun Ku
- Korea Advanced Institute of Science and Technology, Department of Bio and Brain Engineering, Daejeon 34141, Korea
| | - Yujung Kang
- Vieworks Co., R&D Center, Gyeonggi-do 14055, Korea
| | - Chulwoo Ahn
- Yonsei University College of Medicine, Department of Internal Medicine, Seoul 03722, Korea
| | - Chulhee Choi
- Korea Advanced Institute of Science and Technology, Department of Bio and Brain Engineering, Daejeon 34141, KoreabKorea Advanced Institute of Science and Technology, KI for the BioCentury, Daejeon 34141, Korea
| |
Collapse
|
18
|
Kalchenko V, Israeli D, Kuznetsov Y, Meglinski I, Harmelin A. A simple approach for non-invasive transcranial optical vascular imaging (nTOVI). JOURNAL OF BIOPHOTONICS 2015; 8:897-901. [PMID: 25924020 DOI: 10.1002/jbio.201400140] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 02/26/2015] [Accepted: 04/12/2015] [Indexed: 05/21/2023]
Abstract
In vivo imaging of cerebral vasculature is highly vital for clinicians and medical researchers alike. For a number of years non-invasive optical-based imaging of brain vascular network by using standard fluorescence probes has been considered as impossible. In the current paper controverting this paradigm, we present a robust non-invasive optical-based imaging approach that allows visualize major cerebral vessels at the high temporal and spatial resolution. The developed technique is simple to use, utilizes standard fluorescent dyes, inexpensive micro-imaging and computation procedures. The ability to clearly visualize middle cerebral artery and other major vessels of brain vascular network, as well as the measurements of dynamics of blood flow are presented. The developed imaging approach has a great potential in neuroimaging and can significantly expand the capabilities of preclinical functional studies of brain and notably contribute for analysis of cerebral blood circulation in disorder models. An example of 1 × 1.5 cm color-coded image of brain blood vessels of mouse obtained in vivo by transcranial optical vascular imaging (TOVI) approach through the intact cranium.
Collapse
Affiliation(s)
- Vyacheslav Kalchenko
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, 76100, Israel.
| | - David Israeli
- Jerusalem Center for Mental Health, affiliated with the Hebrew University, Jerusalem, Israel
| | - Yuri Kuznetsov
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Igor Meglinski
- Jack Dodd Centre for Quantum Technology, Department of Physics, University of Otago, Dunedin, 9054, New Zealand
- Opto-Electronics and Measurement Techniques Laboratory, University of Oulu, Oulu, FI-9014, Finland
- Interdisciplinary Laboratory of Biophotonics, Tomsk State University, Tomsk, 634050, Russia
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
19
|
Kang HM, Sohn I, Jung J, Jeong JW, Park C. Age-related changes in pial arterial structure and blood flow in mice. Neurobiol Aging 2015; 37:161-170. [PMID: 26460142 DOI: 10.1016/j.neurobiolaging.2015.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
Abstract
Age-related cerebral blood flow decreases are thought to deteriorate cognition and cause senescence, although the related mechanism is unclear. To investigate the relationships between aging and changes in cerebral blood flow and vasculature, we obtained fluorescence images of young (2-month-old) and old (12-month-old) mice using indocyanine green (ICG). First, we found that the blood flow in old mice's brains is lower than that in young mice and that old mice had more curved pial arteries and fewer pial artery junctions than young mice. Second, using Western blotting, we determined that the ratio of collagen to elastin (related to cerebral vascular wall distensibility) increased with age. Finally, we found that the peak ICG intensity and blood flow index decreased, whereas the mean transit time increased, with age in the middle cerebral artery and superior sagittal sinus. Age-related changes in pial arterial structure and composition, concurrent with the observed changes in the blood flow parameters, suggest that age-related changes in the cerebral vasculature structure and distensibility may induce altered brain blood flow.
Collapse
Affiliation(s)
- Hye-Min Kang
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Inkyung Sohn
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Joo-Won Jeong
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Chan Park
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
20
|
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: 1.3] [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
|
21
|
Kang HM, Sohn I, Kim S, Kim D, Jung J, Jeong JW, Park C. Optical measurement of mouse strain differences in cerebral blood flow using indocyanine green. J Cereb Blood Flow Metab 2015; 35:912-6. [PMID: 25833343 PMCID: PMC4640258 DOI: 10.1038/jcbfm.2015.50] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 02/17/2015] [Accepted: 03/04/2015] [Indexed: 11/09/2022]
Abstract
C57BL/6 mice have more cerebral arterial branches and collaterals than BALB/c mice. We measured and compared blood flow dynamics of the middle cerebral artery (MCA) in these two strains, using noninvasive optical imaging with indocyanine green (ICG). Relative maximum fluorescence intensity (Imax) and the time needed for ICG to reach Imax in the MCA of C57BL/c were lower than that in BALB/c mice. Moreover, the mean transit time was significantly lower in C57BL/6 than in BALB/c mice. These data suggest that the higher number of arterial branches and collaterals in C57BL/6 mice yields a lower blood flow per cerebral artery.
Collapse
Affiliation(s)
- Hye-Min Kang
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Inkyung Sohn
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | | | - Daehwan Kim
- R&D Center, Vieworks Co., Ltd., Anyang, Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Joo-Won Jeong
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Chan Park
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
22
|
Kang HM, Sohn I, Park C. Use of indocyanine green for optical analysis of cortical infarcts in photothrombotic ischemic brains. J Neurosci Methods 2015; 248:46-50. [DOI: 10.1016/j.jneumeth.2015.03.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/30/2015] [Accepted: 03/30/2015] [Indexed: 10/23/2022]
|
23
|
Porat Y, Assa-Kunik E, Belkin M, Krakovsky M, Lamensdorf I, Duvdevani R, Sivak G, Niven MJ, Bulvik S. A novel potential therapy for vascular diseases: blood-derived stem/progenitor cells specifically activated by dendritic cells. Diabetes Metab Res Rev 2014; 30:623-34. [PMID: 24638886 DOI: 10.1002/dmrr.2543] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 02/26/2014] [Accepted: 03/03/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND Vascular diseases are a major cause of morbidity and mortality, particularly in diabetic patients. Stem/progenitor cell treatments with bone marrow-derived cells show safety and promising outcomes, albeit not without some preprocedural adverse events related to cell collection and mobilization. We describe a novel technology for generating a therapeutic population (BGC101) of enriched endothelial progenitor cells (EPCs) from non-mobilized blood, using dendritic cells to specifically direct stem/progenitor cell activity in vitro. METHODS AND RESULTS Selected immature plasmacytoid and myeloid dendritic cells from 24 healthy and two diabetic donors were activated with anti-inflammatory and pro-angiogenic molecules to induce specific activation signals. Co-culturing of activated dendritic cells with stem/progenitor cells for 12-66 h generated 83.7 ± 7.4 × 10(6) BGC101 cells with 97% viability from 250 mL of blood. BGC101, comprising 52.4 ± 2.5% EPCs (expressing Ulex-lectin, AcLDL uptake, Tie2, vascular endothelial growth factor receptor 1 and 2, and CD31), 16.1 ± 1.9% stem/progenitor cells (expressing CD34 and CD184) and residual B and T helper cells, demonstrated angiogenic and stemness potential and secretion of interleukin-8, interleukin-10, vascular endothelial growth factor and osteopontin. When administered to immunodeficient mice with limb ischemia (n = 40), BGC101 yielded a high safety profile and significantly increased blood perfusion, capillary density and leg function after 21 days. Cell tracking and biodistribution showed that engraftment was restricted to the ischemic leg. CONCLUSIONS These observations provide preliminary evidence that alternatively activated dendritic cells can promote the generation of EPC-enriched stem/progenitor cells within a 1-day culture. The resulting product BGC101 has the potential for treatment of various vascular conditions such as coronary heart disease, stroke and peripheral ischemia.
Collapse
Affiliation(s)
- Yael Porat
- BioGenCell Ltd, Sanz Medical Center, Laniado Hospital, Netanya, Israel; Sanz Medical Center, Laniado Hospital, Netanya, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Primed 3D injectable microniches enabling low-dosage cell therapy for critical limb ischemia. Proc Natl Acad Sci U S A 2014; 111:13511-6. [PMID: 25197069 DOI: 10.1073/pnas.1411295111] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The promise of cell therapy for repair and restoration of damaged tissues or organs relies on administration of large dose of cells whose healing benefits are still limited and sometimes irreproducible due to uncontrollable cell loss and death at lesion sites. Using a large amount of therapeutic cells increases the costs for cell processing and the risks of side effects. Optimal cell delivery strategies are therefore in urgent need to enhance the specificity, efficacy, and reproducibility of cell therapy leading to minimized cell dosage and side effects. Here, we addressed this unmet need by developing injectable 3D microscale cellular niches (microniches) based on biodegradable gelatin microcryogels (GMs). The microniches are constituted by in vitro priming human adipose-derived mesenchymal stem cells (hMSCs) seeded within GMs resulting in tissue-like ensembles with enriched extracellular matrices and enhanced cell-cell interactions. The primed 3D microniches facilitated cell protection from mechanical insults during injection and in vivo cell retention, survival, and ultimate therapeutic functions in treatment of critical limb ischemia (CLI) in mouse models compared with free cell-based therapy. In particular, 3D microniche-based therapy with 10(5) hMSCs realized better ischemic limb salvage than treatment with 10(6) free-injected hMSCs, the minimum dosage with therapeutic effects for treating CLI in literature. To the best of our knowledge, this is the first convincing demonstration of injectable and primed cell delivery strategy realizing superior therapeutic efficacy for treating CLI with the lowest cell dosage in mouse models. This study offers a widely applicable cell delivery platform technology to boost the healing power of cell regenerative therapy.
Collapse
|
25
|
Hong G, Lee JC, Jha A, Diao S, Nakayama KH, Hou L, Doyle TC, Robinson JT, Antaris AL, Dai H, Cooke JP, Huang NF. Near-infrared II fluorescence for imaging hindlimb vessel regeneration with dynamic tissue perfusion measurement. Circ Cardiovasc Imaging 2014; 7:517-25. [PMID: 24657826 PMCID: PMC4079035 DOI: 10.1161/circimaging.113.000305] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Real-time vascular imaging that provides both anatomic and hemodynamic information could greatly facilitate the diagnosis of vascular diseases and provide accurate assessment of therapeutic effects. Here, we have developed a novel fluorescence-based all-optical method, named near-infrared II (NIR-II) fluorescence imaging, to image murine hindlimb vasculature and blood flow in an experimental model of peripheral arterial disease, by exploiting fluorescence in the NIR-II region (1000-1400 nm) of photon wavelengths. METHODS AND RESULTS Because of the reduced photon scattering of NIR-II fluorescence compared with traditional NIR fluorescence imaging and thus much deeper penetration depth into the body, we demonstrated that the mouse hindlimb vasculature could be imaged with higher spatial resolution than in vivo microscopic computed tomography. Furthermore, imaging during 26 days revealed a significant increase in hindlimb microvascular density in response to experimentally induced ischemia within the first 8 days of the surgery (P<0.005), which was confirmed by histological analysis of microvascular density. Moreover, the tissue perfusion in the ischemic hindlimb could be quantitatively measured by the dynamic NIR-II method, revealing the temporal kinetics of blood flow recovery that resembled microbead-based blood flowmetry and laser Doppler blood spectroscopy. CONCLUSIONS The penetration depth of millimeters, high spatial resolution, and fast acquisition rate of NIR-II imaging make it a useful imaging tool for murine models of vascular disease.
Collapse
Affiliation(s)
- Guosong Hong
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Jerry C Lee
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Arshi Jha
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Shuo Diao
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Karina H Nakayama
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Luqia Hou
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Timothy C Doyle
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Joshua T Robinson
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Alexander L Antaris
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Hongjie Dai
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - John P Cooke
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.)
| | - Ngan F Huang
- From the School of Medicine, Division of Cardiovascular Medicine (J.C.L., A.J., J.P.C., N.F.H.), Department of Pediatrics (T.C.D.), Department of Chemistry (G.H., S.D., J.T.R., A.L.A., H.D.), and Cardiovascular Institute (K.H.N., L.H., H.D., J.P.C., N.F.H.), Stanford University, CA; and Veteran Affairs Palo Alto Health Care System, Palo Alto, CA (K.H.N., L.H., N.F.H.).
| |
Collapse
|
26
|
An Y, Jeon JW, Kwon K, Choi C. Application of dynamic indocyanine green perfusion imaging for evaluation of vasoactive effect of acupuncture: a preliminary follow-up study on normal healthy volunteers. MEDICAL DEVICES-EVIDENCE AND RESEARCH 2014; 7:17-21. [PMID: 24600252 PMCID: PMC3933705 DOI: 10.2147/mder.s56716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Even though acupuncture has long been used for alleviating symptoms related to vascular insufficiency, the clinical effect of acupuncture on peripheral circulation has not been fully confirmed. In this study, we investigated whether a near-infrared optical imaging-based method can be used to evaluate the efficacy of the acupuncture procedure to induce changes in peripheral tissue perfusion. Methods Two normal, healthy controls were treated with acupuncture on two acupoints (LI-4 and SI-3) three times within 1 week. At the first and third visits, participants were examined using indocyanine green (ICG) perfusion imaging before and 10 minutes after the acupuncture procedure. Blood perfusion of the hands was determined after intravenous bolus injection of ICG and dynamic analysis of the fluorescence signals by near-infrared imaging system. Results The blood perfusion rates of the hands were markedly increased immediately after acupuncture at the first trial in both cases. The baseline perfusion rates of the hands measured at the third visit were higher compared to the original basal level in one case; there was no difference in baseline perfusion rates of both hands in another case. In both cases, there was no acute effect of acupuncture on hand perfusion at the third trial. Conclusions These results collectively suggest a potential of the ICG perfusion imaging as an effective evaluation tool to validate the vasoactive effect of acupuncture.
Collapse
Affiliation(s)
- Yuri An
- Department of Bio and Brain Engineering, KAIST, Daejeon, South Korea
| | - Jong Wook Jeon
- Acupuncture, Moxibustion and Meridian Research Centre, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Kihwan Kwon
- Department of Cardiology, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Chulhee Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, South Korea
| |
Collapse
|
27
|
Yoo CH, Na HJ, Lee DS, Heo SC, An Y, Cha J, Choi C, Kim JH, Park JC, Cho YS. Endothelial progenitor cells from human dental pulp-derived iPS cells as a therapeutic target for ischemic vascular diseases. Biomaterials 2013; 34:8149-60. [PMID: 23896001 DOI: 10.1016/j.biomaterials.2013.07.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 07/01/2013] [Indexed: 12/22/2022]
Abstract
Human dental pulp cells (hDPCs) are a valuable source for the generation of patient-specific human induced pluripotent stem cells (hiPSCs). An advanced strategy for the safe and efficient reprogramming of hDPCs and subsequent lineage-specific differentiation is a critical step toward clinical application. In present research, we successfully generated hDPC-iPSCs using only two non-oncogenic factors: Oct4 and Sox2 (2F hDPC-hiPSCs) and evaluated the feasibility of hDPC-iPSCs as substrates for endothelial progenitor cells (EPCs), contributing to EPC-based therapies. Under conventional differentiation conditions, 2F hDPC-hiPSCs showed higher differentiation efficiency, compared to hiPSCs from other cell types, into multipotent CD34(+) EPCs (2F-hEPCs) capable to differentiate into functional endothelial and smooth muscle cells. The angiogenic and neovasculogenic activities of 2F-hEPCs were confirmed using a Matrigel plug assay in mice. In addition, the therapeutic effects of 2F-hEPC transplantation were confirmed in mouse models of hind-limb ischemia and myocardial infarction. Importantly, 2F-EPCs effectively integrated into newly formed vascular structures and enhanced neovascularization via likely both direct and indirect paracrine mechanisms. 2F hDPC-hiPSCs have a robust capability for the generation of angiogenic and vasculogenic EPCs, representing a strategy for patient-specific EPC therapies and disease modeling, particularly for ischemic vascular diseases.
Collapse
Affiliation(s)
- Chae Hwa Yoo
- Stem Cells Research Center, KRIBB, Daejeon, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Anisimov A, Tvorogov D, Alitalo A, Leppänen VM, An Y, Han EC, Orsenigo F, Gaál EI, Holopainen T, Koh YJ, Tammela T, Korpisalo P, Keskitalo S, Jeltsch M, Ylä-Herttuala S, Dejana E, Koh GY, Choi C, Saharinen P, Alitalo K. Vascular endothelial growth factor-angiopoietin chimera with improved properties for therapeutic angiogenesis. Circulation 2013; 127:424-34. [PMID: 23357661 DOI: 10.1161/circulationaha.112.127472] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND There is an unmet need for proangiogenic therapeutic molecules for the treatment of tissue ischemia in cardiovascular diseases. However, major inducers of angiogenesis such as vascular endothelial growth factor (VEGF/VEGF-A) have side effects that limit their therapeutic utility in vivo, especially at high concentrations. Angiopoietin-1 has been considered to be a blood vessel stabilization factor that can inhibit the intrinsic property of VEGF to promote vessel leakiness. In this study, we have designed and tested the angiogenic properties of chimeric molecules consisting of receptor-binding parts of VEGF and angiopoietin-1. We aimed at combining the activities of both factors into 1 molecule for easy delivery and expression in target tissues. METHODS AND RESULTS The VEGF-angiopoietin-1 (VA1) chimeric protein bound to both VEGF receptor-2 and Tie2 and induced the activation of both receptors. Detailed analysis of VA1 versus VEGF revealed differences in the kinetics of VEGF receptor-2 activation and endocytosis, downstream kinase activation, and VE-cadherin internalization. The delivery of a VA1 transgene into mouse skeletal muscle led to increased blood flow and enhanced angiogenesis. VA1 was also very efficient in rescuing ischemic limb perfusion. However, VA1 induced less plasma protein leakage and myeloid inflammatory cell recruitment than VEGF. Furthermore, angioma-like structures associated with VEGF expression were not observed with VA1. CONCLUSIONS The VEGF-angiopoietin-1 chimera is a potent angiogenic factor that triggers a novel mode of VEGF receptor-2 activation, promoting less vessel leakiness, less tissue inflammation, and better perfusion in ischemic muscle than VEGF. These properties of VA1 make it an attractive therapeutic tool.
Collapse
Affiliation(s)
- Andrey Anisimov
- Wihuri Research Institute, Biomedicum Helsinki, PO Box 63 (Haartmaninkatu 8), University of Helsinki, Helsinki, 00014 Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Non-invasive optical imaging techniques, such as fluorescence imaging (FI) or bioluminescence imaging (BLI) have emerged as important tools in biomedical research. As demonstrated in different animal disease models, they enable visualization of physiological and pathophysiological processes at the cellular and molecular level in vivo with high specificity. Optical techniques are easy to use, fast, and affordable. Furthermore, they are characterized by their high sensitivity. In FI, very low amounts of the imaging agent (nano- to femtomol or even less) can be detected. Due to the absorption and scattering of light in tissue, optical techniques exhibit a comparably low spatial resolution in the millimeter range and a depth limit of a few centimeters. However, non-invasive imaging of biological processes in small animals and in outer or inner surfaces as well as during surgery even in humans is feasible. Currently two agents for fluorescence imaging are clinically approved, namely indocyanine green (ICG) and 5-aminolevulinic acid (5-ALA). In the past years, a number of new optical imaging agents for FI and reporter systems for BLI have been developed and successfully tested in animal models. Some of the FI agents might promise the application in clinical oncology. In this chapter, we describe the basic principles of non-invasive optical imaging techniques, give examples for the visualization of biological processes in animal models of cancer, and discuss potential clinical applications in oncology.
Collapse
|
30
|
|
31
|
Göbel W, Brucker D, Kienast Y, Johansson A, Kniebühler G, Rühm A, Eigenbrod S, Fischer S, Goetz M, Kreth FW, Ehrhardt A, Stepp H, Irion KM, Herms J. Optical needle endoscope for safe and precise stereotactically guided biopsy sampling in neurosurgery. OPTICS EXPRESS 2012. [PMID: 23187467 DOI: 10.1364/oe.20.026117] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Proper treatment of deep seated brain tumors requires correct histological diagnosis which unambiguously necessitates biopsy sampling. Stereotactically guided sampling of biopsies is widely used but bears the danger of incorrect sampling locations and damage to intracerebral blood vessels. Here, we present a minimally invasive contact endoscopic probe that can be inserted into the tissue inside a standard biopsy needle and allows for fluorescence detection of both tumorous tissue and intracerebral blood vessels. Outer diameter of our contact probe is smaller than 1.5 mm, field-of-view in the range of several hundred microns; the optical design allows for simultaneous detection and visualization of tissue autofluorescence and selective fluorescence signals from deep seated brain tumors and vasculature as shown on in vivo animal models. We demonstrate the tumor detection capability during stereotactic needle insertion in a clinical pilot trial. Using our probe, we expect stereotactic interventions to become safer and more precise and the technology might ultimately be used also for various other kinds of applications.
Collapse
Affiliation(s)
- Werner Göbel
- Department of Research & Technology, KARL STORZ GmbH & Co. KG, Tuttlingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ku T, Choi C. Noninvasive optical measurement of cerebral blood flow in mice using molecular dynamics analysis of indocyanine green. PLoS One 2012; 7:e48383. [PMID: 23119000 PMCID: PMC3485229 DOI: 10.1371/journal.pone.0048383] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 09/24/2012] [Indexed: 11/18/2022] Open
Abstract
In preclinical studies of ischemic brain disorders, it is crucial to measure cerebral blood flow (CBF); however, this requires radiological techniques with heavy instrumentation or invasive procedures. Here, we propose a noninvasive and easy-to-use optical imaging technique for measuring CBF in experimental small animals. Mice were injected with indocyanine green (ICG) via tail-vein catheterization. Time-series near-infrared fluorescence signals excited by 760 nm light-emitting diodes were imaged overhead by a charge-coupled device coupled with an 830 nm bandpass-filter. We calculated four CBF parameters including arrival time, rising time and mean transit time of a bolus and blood flow index based on time and intensity information of ICG fluorescence dynamics. CBF maps were generated using the parameters to estimate the status of CBF, and they dominantly represented intracerebral blood flows in mice even in the presence of an intact skull and scalp. We demonstrated that this noninvasive optical imaging technique successfully detected reduced local CBF during middle cerebral artery occlusion. We further showed that the proposed method is sufficiently sensitive to detect the differences between CBF status in mice anesthetized with either isoflurane or ketamine–xylazine, and monitor the dynamic changes in CBF after reperfusion during transient middle cerebral artery occlusion. The near-infrared optical imaging of ICG fluorescence combined with a time-series analysis of the molecular dynamics can be a useful noninvasive tool for preclinical studies of brain ischemia.
Collapse
Affiliation(s)
- Taeyun Ku
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Chulhee Choi
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
- KI for the BioCentury, KAIST, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
33
|
A review of indocyanine green fluorescent imaging in surgery. Int J Biomed Imaging 2012; 2012:940585. [PMID: 22577366 PMCID: PMC3346977 DOI: 10.1155/2012/940585] [Citation(s) in RCA: 823] [Impact Index Per Article: 68.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 02/01/2012] [Indexed: 02/07/2023] Open
Abstract
The purpose of this paper is to give an overview of the recent surgical intraoperational applications of indocyanine green fluorescence imaging methods, the basics of the technology, and instrumentation used. Well over 200 papers describing this technique in clinical setting are reviewed. In addition to the surgical applications, other recent medical applications of ICG are briefly examined.
Collapse
|
34
|
Dziekan T, Weissbach C, Voigt J, Ebert B, Macdonald R, Bahner ML, Mahler M, Schirner M, Berliner M, Berliner B, Osel J, Osel I. Detection of rheumatoid arthritis by evaluation of normalized variances of fluorescence time correlation functions. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:076015. [PMID: 21806276 DOI: 10.1117/1.3599958] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Fluorescence imaging using the dye indocyanine green as a contrast agent was investigated in a prospective clinical study for the detection of rheumatoid arthritis. Normalized variances of correlated time series of fluorescence intensities describing the bolus kinetics of the contrast agent in certain regions of interest were analyzed to differentiate healthy from inflamed finger joints. These values are determined using a robust, parameter-free algorithm. We found that the normalized variance of correlation functions improves the differentiation between healthy joints of volunteers and joints with rheumatoid arthritis of patients by about 10% compared to, e.g., ratios of areas under the curves of raw data.
Collapse
Affiliation(s)
- Thomas Dziekan
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Koh YJ, Koh BI, Kim H, Joo HJ, Jin HK, Jeon J, Choi C, Lee DH, Chung JH, Cho CH, Park WS, Ryu JK, Suh JK, Koh GY. Stromal Vascular Fraction From Adipose Tissue Forms Profound Vascular Network Through the Dynamic Reassembly of Blood Endothelial Cells. Arterioscler Thromb Vasc Biol 2011; 31:1141-50. [DOI: 10.1161/atvbaha.110.218206] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective—
Tremendous efforts have been made to establish effective therapeutic neovascularization using adipose tissue-derived stromal vascular fraction (SVF), but the efficiency is low, and underlying mechanisms and their interaction with the host in a new microenvironment are poorly understood.
Methods and Results—
Here we demonstrate that direct implantation of SVF derived from donor adipose tissue can create a profound vascular network through the disassembly and reassembly of blood endothelial cells at the site of implantation. This neovasculature successfully established connection with recipient blood vessels to form a functionally perfused circuit. Addition of vascular growth factors to the SVF implant improved the efficiency of functional neovasculature formation. In contrast, spheroid culture of SVF before implantation reduced the capacity of vasculature formation, possibly because of cellular alteration. Implanting SVF into the mouse ischemic hindlimb induced the robust formation of a local neovascular network and salvaged the limb. Moreover, the coimplantation of SVF prevented fat absorption in the subcutaneous adipose tissue graft model.
Conclusion—
Freshly isolated SVF can effectively induce new vessel formation through the dynamic reassembly of blood endothelial cells and could be applied to achieve therapeutic neovascularization for relieving ischemia and preventing fat absorption in an autologous manner.
Collapse
Affiliation(s)
- Young Jun Koh
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Bong Ihn Koh
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Honsoul Kim
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Hyung Joon Joo
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Ho Kyoung Jin
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Jongwook Jeon
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Chulhee Choi
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Dong Hun Lee
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Jin Ho Chung
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Chung-Hyun Cho
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Won Seok Park
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Ji-Kan Ryu
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Jun Kyu Suh
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Gou Young Koh
- From the Graduate School of Biomedical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea (Y.J.K., B.I.K., H.K., H.J.J., H.K.J., J.J., C.C., G.Y.K.); Institute of Dermatological Science, Medical Research Center (D.H.L., J.H.C.), and Department of Pharmacology (C.-H.C.), Seoul National University College of Medicine, Seoul, Korea, R&D Center, Amore Pacific, Gyeonggi, Korea (W.S.P.); Department of Urology, Inha University School of Medicine, Incheon, Korea
| |
Collapse
|
36
|
Min JK, Park H, Choi HJ, Kim Y, Pyun BJ, Agrawal V, Song BW, Jeon J, Maeng YS, Rho SS, Shim S, Chai JH, Koo BK, Hong HJ, Yun CO, Choi C, Kim YM, Hwang KC, Kwon YG. The WNT antagonist Dickkopf2 promotes angiogenesis in rodent and human endothelial cells. J Clin Invest 2011; 121:1882-93. [PMID: 21540552 DOI: 10.1172/jci42556] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 02/16/2011] [Indexed: 12/23/2022] Open
Abstract
Neovessel formation is a complex process governed by the orchestrated action of multiple factors that regulate EC specification and dynamics within a growing vascular tree. These factors have been widely exploited to develop therapies for angiogenesis-related diseases such as diabetic retinopathy and tumor growth and metastasis. WNT signaling has been implicated in the regulation and development of the vascular system, but the detailed mechanism of this process remains unclear. Here, we report that Dickkopf1 (DKK1) and Dickkopf2 (DKK2), originally known as WNT antagonists, play opposite functional roles in regulating angiogenesis. DKK2 induced during EC morphogenesis promoted angiogenesis in cultured human endothelial cells and in in vivo assays using mice. Its structural homolog, DKK1, suppressed angiogenesis and was repressed upon induction of morphogenesis. Importantly, local injection of DKK2 protein significantly improved tissue repair, with enhanced neovascularization in animal models of both hind limb ischemia and myocardial infarction. We further showed that DKK2 stimulated filopodial dynamics and angiogenic sprouting of ECs via a signaling cascade involving LRP6-mediated APC/Asef2/Cdc42 activation. Thus, our findings demonstrate the distinct functions of DKK1 and DKK2 in controlling angiogenesis and suggest that DKK2 may be a viable therapeutic target in the treatment of ischemic vascular diseases.
Collapse
Affiliation(s)
- Jeong-Ki Min
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kang Y, Lee J, An Y, Jeon J, Choi C. Segmental analysis of indocyanine green pharmacokinetics for the reliable diagnosis of functional vascular insufficiency. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:030504. [PMID: 21456859 DOI: 10.1117/1.3556718] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Accurate and reliable diagnosis of functional insufficiency of peripheral vasculature is essential since Raynaud phenomenon (RP), most common form of peripheral vascular insufficiency, is commonly associated with systemic vascular disorders. We have previously demonstrated that dynamic imaging of near-infrared fluorophore indocyanine green (ICG) can be a noninvasive and sensitive tool to measure tissue perfusion. In the present study, we demonstrated that combined analysis of multiple parameters, especially onset time and modified T(max) which means the time from onset of ICG fluorescence to T(max), can be used as a reliable diagnostic tool for RP. To validate the method, we performed the conventional thermographic analysis combined with cold challenge and rewarming along with ICG dynamic imaging and segmental analysis. A case-control analysis demonstrated that segmental pattern of ICG dynamics in both hands was significantly different between normal and RP case, suggesting the possibility of clinical application of this novel method for the convenient and reliable diagnosis of RP.
Collapse
|
38
|
Aqueous extract of the medicinal plant Patrinia villosa Juss. Induces angiogenesis via activation of focal adhesion kinase. Microvasc Res 2010; 80:303-9. [DOI: 10.1016/j.mvr.2010.05.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 05/23/2010] [Accepted: 05/25/2010] [Indexed: 11/30/2022]
|
39
|
Kang Y, Lee J, Kwon K, Choi C. Application of novel dynamic optical imaging for evaluation of peripheral tissue perfusion. Int J Cardiol 2010; 145:e99-101. [PMID: 19230993 DOI: 10.1016/j.ijcard.2008.12.166] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 12/22/2008] [Indexed: 10/21/2022]
|
40
|
Efficient differentiation of human pluripotent stem cells into functional CD34+ progenitor cells by combined modulation of the MEK/ERK and BMP4 signaling pathways. Blood 2010; 116:5762-72. [PMID: 20884805 DOI: 10.1182/blood-2010-04-280719] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Differentiation of human pluripotent stem cells (hPSCs) into functional cell types is a crucial step in cell therapy. In the present study, we demonstrate that functional CD34(+) progenitor cells can be efficiently produced from human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) by combined modulation of 2 signaling pathways. A higher proportion of CD34(+) cells (∼ 20%) could be derived from hPSCs by inhibition of mitogen-activated protein kinase (MAPK) extracellular signal-regulated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling and activation of bone morphogenic protein-4 (BMP4) signaling. hPSC-derived CD34(+) progenitor cells further developed to endothelial and smooth muscle cells with functionality. Moreover, they contributed directly to neovasculogenesis in ischemic mouse hind limbs, thereby resulting in improved blood perfusion and limb salvage. Our results suggest that combined modulation of signaling pathways may be an efficient means of differentiating hPSCs into functional CD34(+) progenitor cells.
Collapse
|