1
|
van Boxtel J, Uguten M, Harmsen MC, Stevens HP, van Dongen JA. Isolation of Stromal Vascular Fraction by Fractionation of Adipose Tissue. Methods Mol Biol 2025; 2922:97-111. [PMID: 40208530 DOI: 10.1007/978-1-0716-4510-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Adipose tissue-derived stromal cells (ASCs) are promising candidates for cellular therapy in the field of regenerative medicine. ASCs are multipotent mesenchymal stem cell-like and reside in the stromal vascular fraction (SVF) of adipose tissue with the capacity to secrete a plethora of pro-regenerative growth factors. Future applications of ASCs may be restricted through (trans)national governmental policies that do not allow for use of non-human-derived (non-autologous) enzymes to isolate ASC. Besides, enzymatic isolation procedures are also time-consuming. To overcome this issue, non-enzymatic isolation procedures to isolate ASCs or the SVF have been developed, such as the fractionation of adipose tissue procedure (FAT). This standardized procedure to isolate the stromal vascular fraction can be performed within 10-12 min. The short procedure time allows for intra-operative isolation of 1 mL of stromal vascular fraction derived from 10 mL of centrifuged adipose tissue. The stromal vascular fraction mostly contains blood vessels, extracellular matrix, and ASCs. However, based on the histological stainings, an interdonor variation exists which might result in different therapeutic effects. The existing interdonor variations can be addressed by histological stainings and flow cytometry. Furthermore, the re-usable open system has been replaced by a validated disposable semi-closed system with a one-hole fractionator.
Collapse
Affiliation(s)
- Joeri van Boxtel
- Department of Plastic, Reconstuctive and Hand Surgery, Catharina Hospital Eindhoven, Eindhoven, The Netherlands
| | - Mustafa Uguten
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Martin C Harmsen
- Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hieronymus P Stevens
- PRSkliniek, Vlaardingen, The Netherlands
- Department of Aesthetic Surgery, Velthuis Clinics, Rotterdam, The Netherlands
| | - Joris A van Dongen
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
2
|
Kavaliunaite E, Dhumale P, Jensen CH, Sheikh SP, Lindholt JS, Stubbe J. A Single Injection of ADRCs Does Not Prevent AAA Formation in Rats in a Randomized Blinded Design. Int J Mol Sci 2024; 25:7591. [PMID: 39062833 PMCID: PMC11276694 DOI: 10.3390/ijms25147591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
There is a pressing need for alternative medical treatments for abdominal aortic aneurysms (AAAs). Mesenchymal regenerative cells derived from adipose tissue (ADRCs) have shown potential in modulating the inflammation and immune responses that drive AAA progression. We hypothesized that ADRCs could reduce inflammation and preserve vascular integrity, potentially slowing the progression of AAA. In our study, subcutaneous adipose tissue was harvested from male Sprague Dawley rats, from which ADRCs were isolated. AAA was induced in these rats using intraluminal porcine pancreatic elastase, followed by intravenous administration of either ADRCs (106 cells) or saline (0.1 mL). We monitored the progression of AAA through weekly ultrasound, and the rats were sacrificed on day 28 for histological analysis. Our results showed no significant difference in the inner abdominal aortic diameter at day 28 between the control group (172% ± 73%, n = 17) and the ADRC-treated group (181% ± 75%, n = 15). Histological analyses of AAA cross-sections also revealed no significant difference in the infiltration of neutrophils or macrophages between the two groups. Furthermore, the integrity and content of elastin in the tunica media were similar between groups. These findings indicate that a single injection of ADRCs does not inhibit the development of AAA in rats in a randomized blinded study.
Collapse
Affiliation(s)
- Egle Kavaliunaite
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Cardiac, Thoracic and Vascular Surgery, Odense University Hospital (OUH), 5000 Odense C, Denmark;
| | - Pratibha Dhumale
- Department of Clinical Research, University of Southern Denmark (SDU), 5230 Odense M, Denmark; (P.D.)
- Department of Clinical Biochemistry, Odense University Hospital (OUH), 5000 Odense C, Denmark
| | - Charlotte Harken Jensen
- Department of Clinical Research, University of Southern Denmark (SDU), 5230 Odense M, Denmark; (P.D.)
- Department of Clinical Biochemistry, Odense University Hospital (OUH), 5000 Odense C, Denmark
| | - Søren P. Sheikh
- Open Patient Data Explorative Network, Institute of Clinical Research, Odense University Hospital (OUH), 5000 Odense C, Denmark
| | - Jes S. Lindholt
- Department of Cardiac, Thoracic and Vascular Surgery, Odense University Hospital (OUH), 5000 Odense C, Denmark;
| | - Jane Stubbe
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
| |
Collapse
|
3
|
Akerman AW, Alexander KC, Caranasos TG, Ikonomidis JS. Therapeutic potential of mesenchymal stem cells and their secreted extracellular vesicles in thoracic aortic aneurysm disease. J Thorac Cardiovasc Surg 2024; 167:89-93.e1. [PMID: 37084818 PMCID: PMC10882625 DOI: 10.1016/j.jtcvs.2023.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/23/2023]
Affiliation(s)
- Adam W Akerman
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Kyle C Alexander
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Thomas G Caranasos
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - John S Ikonomidis
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, NC.
| |
Collapse
|
4
|
Aronowitz JA, Winterhalter B. Adipose-Derived Regenerative Cellular Therapy of Chronic Wounds. Regen Med 2023. [DOI: 10.1007/978-3-030-75517-1_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
5
|
Immunomodulation of Skin Repair: Cell-Based Therapeutic Strategies for Skin Replacement (A Comprehensive Review). Biomedicines 2022; 10:biomedicines10010118. [PMID: 35052797 PMCID: PMC8773777 DOI: 10.3390/biomedicines10010118] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
The immune system has a crucial role in skin wound healing and the application of specific cell-laden immunomodulating biomaterials emerged as a possible treatment option to drive skin tissue regeneration. Cell-laden tissue-engineered skin substitutes have the ability to activate immune pathways, even in the absence of other immune-stimulating signals. In particular, mesenchymal stem cells with their immunomodulatory properties can create a specific immune microenvironment to reduce inflammation, scarring, and support skin regeneration. This review presents an overview of current wound care techniques including skin tissue engineering and biomaterials as a novel and promising approach. We highlight the plasticity and different roles of immune cells, in particular macrophages during various stages of skin wound healing. These aspects are pivotal to promote the regeneration of nonhealing wounds such as ulcers in diabetic patients. We believe that a better understanding of the intrinsic immunomodulatory features of stem cells in implantable skin substitutes will lead to new translational opportunities. This, in turn, will improve skin tissue engineering and regenerative medicine applications.
Collapse
|
6
|
LncRNA Sox2ot modulates the progression of thoracic aortic aneurysm by regulating miR-330-5p/Myh11. Biosci Rep 2021; 40:225469. [PMID: 32578852 PMCID: PMC7364482 DOI: 10.1042/bsr20194040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/21/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022] Open
Abstract
Thoracic aortic aneurysm (TAA) has been causing the death of elder people. Myosin heavy chain 11 (Myh11) has been reported associated with aortic aneurysm, but there is no specific study on its function on TAA. Here we aimed to explore the function of Myh11 on mouse aortic smooth muscle cells (SMCs) for studying the inner mechanism of TAA. H2O2 treatment was implemented on mouse aortic SMCs for detecting cell apoptosis. Meanwhile, functional assays were conducted to verify the function of Myh11 on mouse aortic SMCs. Also, pull-down assay, RIP assay were implemented to identify the potential RNAs for study. Quantitative real-time polymerase chain reaction (qRT-PCR) and luciferase reporter assay were implemented to identify the expression and binding relationships of RNAs. Myh11 expression was increased by treatment of H2O2. Myh11 could decrease proliferation and enhance apoptosis of mouse aortic SMCs. At the same time, mmu-miR-330-5p could bind to Myh11 and Sox2ot, forming a competing endogenous RNA (ceRNA) pathway to regulate the proliferation and apoptosis of mouse aortic SMCs. Moreover, both Sox2ot and Myh11 were proved to be up-regulated whereas miR-330-5p down-regulated in Fbn1C1039G/+ mice, the in vivo model of TAA. In a word, long noncoding RNA (lncRNA) Sox2ot modulates the progression of TAA by regulating miR-330-5p/Myh11 axis.
Collapse
|
7
|
Ye T, Zhang G, Liu H, Shi J, Qiu H, Liu Y, Han F, Hou N. Relationships Between Perivascular Adipose Tissue and Abdominal Aortic Aneurysms. Front Endocrinol (Lausanne) 2021; 12:704845. [PMID: 34194399 PMCID: PMC8236981 DOI: 10.3389/fendo.2021.704845] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) are typically asymptomatic, and there is a high mortality rate associated with aneurysm rupture. AAA pathogenesis involves extracellular matrix degradation, vascular smooth muscle cell phenotype switching, inflammation, and oxidative stress. There is increasing evidence of excessive adipocyte accumulation in ruptured AAA walls. These excessive numbers of adipocytes in the vascular wall have been closely linked with AAA progression. Perivascular adipose tissue (PVAT), a unique type of adipose tissue, can be involved in adipocyte accumulation in the AAA wall. PVAT produces various chemokines and adipocytokines around vessels to maintain vascular homeostasis through paracrine and autocrine mechanisms in normal physiological conditions. Nevertheless, PVAT loses its normal function and promotes the progression of vascular diseases in pathological conditions. There is evidence of significantly reduced AAA diameter in vessel walls of removed PVAT. There is a need to highlight the critical roles of cytokines, cells, and microRNA derived from PVAT in the regulation of AAA development. PVAT may constitute an important therapeutic target for the prevention and treatment of AAAs. In this review, we discuss the relationship between PVAT and AAA development; we also highlight the potential for PVAT-derived factors to serve as a therapeutic target in the treatment of AAAs.
Collapse
Affiliation(s)
- Tongtong Ye
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Guangdong Zhang
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hangyu Liu
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, China
| | - Junfeng Shi
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongyan Qiu
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yongping Liu
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Ningning Hou, ; Fang Han,
| | - Ningning Hou
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Ningning Hou, ; Fang Han,
| |
Collapse
|
8
|
Parvizi M, Ryan ZC, Ebtehaj S, Arendt BK, Lanza IR. The secretome of senescent preadipocytes influences the phenotype and function of cells of the vascular wall. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165983. [PMID: 33002577 DOI: 10.1016/j.bbadis.2020.165983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/16/2020] [Accepted: 09/24/2020] [Indexed: 01/10/2023]
Abstract
Senescent cells accumulate in numerous tissues in several chronic conditions such as aging, obesity, and diabetes. These cells are in a state of irreversible cell-cycle arrest and secrete inflammatory cytokines, chemokines and other immune modulators that have paracrine effects on nearby tissues. Adipose tissue, in particular, harbors senescent cells, which have been linked with numerous chronic conditions and age-related comorbidities. Here we performed a series of in vitro experiments to determine the influence of senescent preadipocytes on key cell types found in vessel walls, including vascular smooth muscle cells (VSMCs), endothelial cells (ECs), macrophages (MQs), and adipose-derived stromal/stem cells (ASCs). Primary human preadipocytes were irradiated to trigger a senescence-like phenotype. VSMCs, ECs, MQs, and ASCs were exposed to conditioned media collected from irradiated preadipocytes or control preadipocytes. Additional experiments were performed where VSMCs, ECs, MQs, and ASCs were co-cultured with irradiated or control preadipocytes. The secretome of irradiated cells induced an inflammatory phenotype, decreased cell viability, disrupted proliferation and migration, and impaired metabolic function of these cell types in vitro. These maladaptive changes in response to senescent cell exposure provide early evidence in support of a hypothesis that senescent preadipocytes trigger phenotypic and functional changes in key cellular components of blood vessels that may contribute to vascular disease.
Collapse
Affiliation(s)
- Mojtaba Parvizi
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Zachary C Ryan
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Sanam Ebtehaj
- Department of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Bonnie K Arendt
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Ian R Lanza
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
9
|
Wen H, Wang M, Gong S, Li X, Meng J, Wen J, Wang Y, Zhang S, Xin S. Human Umbilical Cord Mesenchymal Stem Cells Attenuate Abdominal Aortic Aneurysm Progression in Sprague-Dawley Rats: Implication of Vascular Smooth Muscle Cell Phenotypic Modulation. Stem Cells Dev 2020; 29:981-993. [PMID: 32486904 PMCID: PMC7410303 DOI: 10.1089/scd.2020.0058] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is life-threatening, for which efficient nonsurgical treatment strategy has not been available so far. Several previous studies investigating the therapeutic effect of mesenchymal stem cells (MSCs) in AAA indicated that MSCs could inhibit aneurysmal inflammatory responses and extracellular matrix destruction, and suppress aneurysm occurrence and expansion. Vascular smooth muscle cell (VSMC) phenotypic plasticity is reported to be predisposed in AAA initiation and progression. However, little is known about the effect of MSCs on VSMC phenotypic modulation in AAA. In this study, we investigate the therapeutic efficacy of umbilical cord mesenchymal stem cells (UC-MSCs) in elastase-induced AAA model and evaluate the effect of UC-MSC on VSMC phenotypic regulation. We demonstrate that the intravenous injection of UC-MSC attenuates elastase-induced aneurysmal expansion, reduces elastin degradation and fragmentation, inhibits MMPs and TNF-α expression, and preserves and/or restores VSMC contractile phenotype in AAA. Taken together, these results highlight the therapeutic and VSMC phenotypic modulation effects of UC-MSC in AAA progression, which further indicates the potential of applying UC-MSC as an alternative treatment candidate for AAA.
Collapse
Affiliation(s)
- Hao Wen
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China.,Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Mingjing Wang
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Shiqiang Gong
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Xintong Li
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China.,Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Jinze Meng
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Jie Wen
- Department of Ultrasonography, Inner Mongolia Baotou City Central Hospital, Baotou, China
| | - Yifei Wang
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Shuqing Zhang
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Shijie Xin
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China.,Regenerative Medicine Research Center of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Ni H, Zhao Y, Ji Y, Shen J, Xiang M, Xie Y. Adipose-derived stem cells contribute to cardiovascular remodeling. Aging (Albany NY) 2019; 11:11756-11769. [PMID: 31800397 PMCID: PMC6932876 DOI: 10.18632/aging.102491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/17/2019] [Indexed: 02/06/2023]
Abstract
Obesity is an independent risk factor for cardiovascular disease. Adipose tissue was initially thought to be involved in metabolism through paracrine. Recent researches discovered mesenchymal stem cells inside adipose tissue which could differentiate into vascular lineages in vitro and in vivo, participating vascular remodeling. However, there were few researches focusing on distinct characteristics and functions of adipose-derived stem cells (ADSCs) from different regions. This is the first comprehensive review demonstrating the variances of ADSCs from the perspective of their origins.
Collapse
Affiliation(s)
- Hui Ni
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiming Zhao
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongli Ji
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Zhang Z, Zou G, Chen X, Lu W, Liu J, Zhai S, Qiao G. Knockdown of lncRNA PVT1 Inhibits Vascular Smooth Muscle Cell Apoptosis and Extracellular Matrix Disruption in a Murine Abdominal Aortic Aneurysm Model. Mol Cells 2019; 42:218-227. [PMID: 30726659 PMCID: PMC6449717 DOI: 10.14348/molcells.2018.0162] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 10/08/2018] [Accepted: 10/15/2018] [Indexed: 01/08/2023] Open
Abstract
This study was designed to determine the effects of the long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) on vascular smooth muscle cell (VSMC) apoptosis and extracellular matrix (ECM) disruption in a murine abdominal aortic aneurysm (AAA) model. After injection of PVT1-silencing lentiviruses, AAA was induced in Apolipoprotein E-deficient (ApoE-/-) male mice by angiotensin II (Ang II) infusion for four weeks. After Ang II infusion, mouse serum levels of pro-inflammatory cytokines were analysed, and aortic tissues were isolated for histological, RNA, and protein analysis. Our results also showed that PVT1 expression was significantly upregulated in abdominal aortic tissues from AAA patients compared with that in controls. Additionally, Ang II treatment significantly increased PVT1 expression, both in cultured mouse VSMCs and in AAA murine abdominal aortic tissues. Of note, the effects of Ang II in facilitating cell apoptosis, increasing matrix metalloproteinase (MMP)-2 and MMP-9, reducing tissue inhibitor of MMP (TIMP)-1, and promoting switching from the contractile to synthetic phenotype in cultured VSMCs were enhanced by overexpression of PVT1 but attenuated by knockdown of PVT1. Furthermore, knockdown of PVT1 reversed Ang II-induced AAA-associated alterations in mice, as evidenced by attenuation of aortic diameter dilation, marked adventitial thickening, loss of elastin in the aorta, enhanced aortic cell apoptosis, elevated MMP-2 and MMP-9, reduced TIMP-1, and increased pro-inflammatory cytokines. In conclusion, our findings demonstrate that knockdown of lncRNA PVT1 suppresses VSMC apoptosis, ECM disruption, and serum pro-inflammatory cytokines in a murine Ang II-induced AAA model.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Angiotensin II/pharmacology
- Animals
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/pathology
- Apolipoproteins E/deficiency
- Apoptosis/drug effects
- Disease Models, Animal
- Extracellular Matrix/metabolism
- Gene Knockdown Techniques
- Humans
- Inflammation/pathology
- Mice
- Middle Aged
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Small Interfering/metabolism
Collapse
Affiliation(s)
- Zhidong Zhang
- Department of Vascular and Endovascular Surgery, Henan Provincial People’s Hospital, Henan,
China
- Department of Aortic Surgery, Fuwai Central China Cardiovascular Hospital, Henan,
China
| | - Gangqiang Zou
- Department of Vascular and Endovascular Surgery, Henan Provincial People’s Hospital, Henan,
China
- Department of Aortic Surgery, Fuwai Central China Cardiovascular Hospital, Henan,
China
| | - Xiaosan Chen
- Department of Vascular and Endovascular Surgery, Henan Provincial People’s Hospital, Henan,
China
- Department of Aortic Surgery, Fuwai Central China Cardiovascular Hospital, Henan,
China
| | - Wei Lu
- Department of Vascular and Endovascular Surgery, Henan Provincial People’s Hospital, Henan,
China
- Department of Aortic Surgery, Fuwai Central China Cardiovascular Hospital, Henan,
China
| | - Jianyang Liu
- Department of Vascular and Endovascular Surgery, Henan Provincial People’s Hospital, Henan,
China
- Department of Aortic Surgery, Fuwai Central China Cardiovascular Hospital, Henan,
China
| | - Shuiting Zhai
- Department of Vascular and Endovascular Surgery, Henan Provincial People’s Hospital, Henan,
China
- Department of Vascular and Endovascular Surgery, Fuwai Central China Cardiovascular Hospital, Henan,
China
| | - Gang Qiao
- Department of Vascular and Endovascular Surgery, Henan Provincial People’s Hospital, Henan,
China
- Department of Aortic Surgery, Fuwai Central China Cardiovascular Hospital, Henan,
China
| |
Collapse
|
12
|
van Dongen JA, Harmsen MC, Stevens HP. Isolation of Stromal Vascular Fraction by Fractionation of Adipose Tissue. Methods Mol Biol 2019; 1993:91-103. [PMID: 31148081 DOI: 10.1007/978-1-4939-9473-1_8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adipose tissue-derived stromal cells (ASCs) are a promising candidates for cellular therapy in the field of regenerative medicine. ASCs are multipotent mesenchymal stem cell-like and reside in the stromal vascular fraction (SVF) of adipose tissue with the capacity to secrete a plethora of pro-regenerative growth factors. Future applications of ASCs may be restricted through (trans)national governmental policies that do not allow for use of nonhuman-derived (non-autologous) enzymes to isolate ASC. Besides, enzymatic isolation procedures are also time consuming. To overcome this issue, nonenzymatic isolation procedures to isolate ASCs or the SVF are being developed, such as the fractionation of adipose tissue procedure (FAT). This standardized procedure to isolate the stromal vascular fraction can be performed within 10-12 min. The short procedure time allows for intraoperative isolation of 1 mL of stromal vascular fraction derived from 10 mL of centrifuged adipose tissue. The stromal vascular fraction mostly contains blood vessels, extracellular matrix, and ASCs. However, based on the histological stainings an interdonor variation exists which might result in different therapeutic effects. The existing interdonor variations can be addressed by histological stainings and flow cytometry.
Collapse
Affiliation(s)
- Joris A van Dongen
- Plastic Surgery Department, Velthuis Kliniek, 3062 MB,, K.P. van der Mandelelaan 10, Rotterdam, The Netherlands
- Department of Pathology and Medical Biology, University of Groningen and University Medical Centre of Groningen, Groningen, The Netherlands
| | - Martin C Harmsen
- Department of Pathology and Medical Biology, University of Groningen and University Medical Centre of Groningen, Groningen, The Netherlands
| | - Hieronymus P Stevens
- Plastic Surgery Department, Velthuis Kliniek, 3062 MB,, K.P. van der Mandelelaan 10, Rotterdam, The Netherlands
| |
Collapse
|
13
|
Augmentation of Dermal Wound Healing by Adipose Tissue-Derived Stromal Cells (ASC). Bioengineering (Basel) 2018; 5:bioengineering5040091. [PMID: 30373121 PMCID: PMC6316823 DOI: 10.3390/bioengineering5040091] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/21/2018] [Accepted: 10/24/2018] [Indexed: 12/14/2022] Open
Abstract
The skin is the largest organ of the human body and is the first line of defense against physical and biological damage. Thus, the skin is equipped to self-repair and regenerates after trauma. Skin regeneration after damage comprises a tightly spatial-temporally regulated process of wound healing that involves virtually all cell types in the skin. Wound healing features five partially overlapping stages: homeostasis, inflammation, proliferation, re-epithelization, and finally resolution or fibrosis. Dysreguled wound healing may resolve in dermal scarring. Adipose tissue is long known for its suppressive influence on dermal scarring. Cultured adipose tissue-derived stromal cells (ASCs) secrete a plethora of regenerative growth factors and immune mediators that influence processes during wound healing e.g., angiogenesis, modulation of inflammation and extracellular matrix remodeling. In clinical practice, ASCs are usually administered as part of fractionated adipose tissue i.e., as part of enzymatically isolated SVF (cellular SVF), mechanically isolated SVF (tissue SVF), or as lipograft. Enzymatic isolation of SVF obtained adipose tissue results in suspension of adipocyte-free cells (cSVF) that lack intact intercellular adhesions or connections to extracellular matrix (ECM). Mechanical isolation of SVF from adipose tissue destructs the parenchyma (adipocytes), which results in a tissue SVF (tSVF) with intact connections between cells, as well as matrix. To date, due to a lack of well-designed prospective randomized clinical trials, neither cSVF, tSVF, whole adipose tissue, or cultured ASCs can be indicated as the preferred preparation procedure prior to therapeutic administration. In this review, we present and discuss current literature regarding the different administration options to apply ASCs (i.e., cultured ASCs, cSVF, tSVF, and lipografting) to augment dermal wound healing, as well as the available indications for clinical efficacy.
Collapse
|
14
|
Parvizi M, Petersen AH, van Spreuwel-Goossens CAFM, Kluijtmans SGJM, Harmsen MC. Perivascular scaffolds loaded with adipose tissue-derived stromal cells attenuate development and progression of abdominal aortic aneurysm in rats. J Biomed Mater Res A 2018; 106:2494-2506. [DOI: 10.1002/jbm.a.36445] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/05/2018] [Accepted: 04/05/2018] [Indexed: 12/19/2022]
Affiliation(s)
- M. Parvizi
- Department of Pathology and Medical Biology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - A. H. Petersen
- Department of Pathology and Medical Biology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | | | | | - M. C. Harmsen
- Department of Pathology and Medical Biology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
15
|
Spinosa M, Lu G, Su G, Bontha SV, Gehrau R, Salmon MD, Smith JR, Weiss ML, Mas VR, Upchurch GR, Sharma AK. Human mesenchymal stromal cell-derived extracellular vesicles attenuate aortic aneurysm formation and macrophage activation via microRNA-147. FASEB J 2018; 32:fj201701138RR. [PMID: 29812968 PMCID: PMC6181641 DOI: 10.1096/fj.201701138rr] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
The formation of an abdominal aortic aneurysm (AAA) is characterized by inflammation, macrophage infiltration, and vascular remodeling. In this study, we tested the hypothesis that mesenchymal stromal cell (MSC)-derived extracellular vesicles (EVs) immunomodulate aortic inflammation, to mitigate AAA formation via modulation of microRNA-147. An elastase-treatment model of AAA was used in male C57BL/6 wild-type (WT) mice. Administration of EVs in elastase-treated WT mice caused a significant attenuation of aortic diameter and mitigated proinflammatory cytokines, inflammatory cell infiltration, an increase in smooth muscle cell α-actin expression, and a decrease in elastic fiber disruption, compared with untreated mice. A 10-fold up-regulation of microRNA (miR)-147, a key mediator of macrophage inflammatory responses, was observed in murine aortic tissue in elastase-treated mice compared with controls on d 14. EVs derived from MSCs transfected with miR-147 mimic, but not with miR-147 inhibitor, attenuated aortic diameter, inflammation, and leukocyte infiltration in elastase-treated mice. In vitro studies of human aortic tissue explants and murine-derived CD11b+ macrophages induced proinflammatory cytokines after elastase treatment, and the expression was attenuated by cocultures with EVs transfected with miR-147 mimic, but not with miR-147 inhibitor. Thus, our findings define a critical role of MSC-derived EVs in attenuation of aortic inflammation and macrophage activation via miR-147 during AAA formation.-Spinosa, M., Lu, G., Su, G., Bontha, S. V., Gehrau, R., Salmon, M. D., Smith, J. R., Weiss, M. L., Mas, V. R., Upchurch, G. R., Sharma, A. K. Human mesenchymal stromal cell-derived extracellular vesicles attenuate aortic aneurysm formation and macrophage activation via microRNA-147.
Collapse
Affiliation(s)
- Michael Spinosa
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Guanyi Lu
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Gang Su
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Sai Vineela Bontha
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Ricardo Gehrau
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Morgan D. Salmon
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Joseph R. Smith
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, USA
| | - Mark L. Weiss
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, USA
| | - Valeria R. Mas
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Gilbert R. Upchurch
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Ashish K. Sharma
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
16
|
Yamawaki-Ogata A, Oshima H, Usui A, Narita Y. Bone marrow–derived mesenchymal stromal cells regress aortic aneurysm via the NF-kB, Smad3 and Akt signaling pathways. Cytotherapy 2017; 19:1167-1175. [DOI: 10.1016/j.jcyt.2017.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/19/2017] [Accepted: 07/28/2017] [Indexed: 12/11/2022]
|
17
|
van Dongen JA, Tuin AJ, Spiekman M, Jansma J, van der Lei B, Harmsen MC. Comparison of intraoperative procedures for isolation of clinical grade stromal vascular fraction for regenerative purposes: a systematic review. J Tissue Eng Regen Med 2017; 12:e261-e274. [PMID: 28084666 DOI: 10.1002/term.2407] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 11/11/2016] [Accepted: 01/09/2017] [Indexed: 12/16/2022]
Abstract
Intraoperative application of the stromal vascular fraction (SVF) of adipose tissue requires a fast and efficient isolation procedure of adipose tissue. This review was performed to systematically assess and compare procedures currently used for the intraoperative isolation of cellular SVF (cSVF) and tissue SVF (tSVF) that still contain the extracellular matrix. Pubmed, EMBASE and the Cochrane central register of controlled trials databases were searched for studies that compare procedures for intraoperative isolation of SVF (searched 28 September 2016). Outcomes of interest were cell yield, viability of cells, composition of SVF, duration, cost and procedure characteristics. Procedures were subdivided into procedures resulting in a cSVF or tSVF. Thirteen out of 3038 studies, evaluating 18 intraoperative isolation procedures, were considered eligible. In general, cSVF and tSVF intraoperative isolation procedures had similar cell yield, cell viability and SVF composition compared to a nonintraoperative (i.e. culture laboratory-based collagenase protocol) control group within the same studies. The majority of intraoperative isolation procedures are less time consuming than nonintraoperative control groups, however. Intraoperative isolation procedures are less time-consuming than nonintraoperative control groups with similar cell yield, viability of cells and composition of SVF, and therefore more suitable for use in the clinic. Nevertheless, none of the intraoperative isolation procedures could be designated as the preferred procedure to isolate SVF. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Joris A van Dongen
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands.,Department of Plastic Surgery, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - A Jorien Tuin
- Department of Oral & Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Maroesjka Spiekman
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Johan Jansma
- Department of Oral & Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Berend van der Lei
- Department of Plastic Surgery, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands.,Bergman Clinics, locations Heerenveen, Zwolle and Groningen, the Netherlands
| | - Martin C Harmsen
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
18
|
Gonzalez-Garza MT, Cruz-Vega DE. Regenerative capacity of autologous stem cell transplantation in elderly: a report of biomedical outcomes. Regen Med 2017; 12:169-178. [DOI: 10.2217/rme-2016-0038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The occurrence of chronic diseases such as neurological, metabolic and cardiovascular degenerative disorders increases with age. Cell therapy is an emerging approach to the treatment of these conditions. Of particular interest is the application of autologous stem cells because it eliminates post-transplantation immune rejection and there are less ethical concerns associated with their use. The regenerative capacity of stem cells harvested from elderly people is however controversial. In this review, we analyze if self-renewal potential, differentiation capability and expression of stemness genes in stem cells collected from elderly patients validate their application in clinical trials and examine the results.
Collapse
Affiliation(s)
| | - Delia Elva Cruz-Vega
- Tecnologico de Monterrey, Escuela Nacional de Medicina, Morones Prieto 3000 Pte, CP64710, Monterrey, Mexico
| |
Collapse
|
19
|
Shen YH, LeMaire SA. Molecular pathogenesis of genetic and sporadic aortic aneurysms and dissections. Curr Probl Surg 2017; 54:95-155. [PMID: 28521856 DOI: 10.1067/j.cpsurg.2017.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX.
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
20
|
Cao Y, Gang X, Sun C, Wang G. Mesenchymal Stem Cells Improve Healing of Diabetic Foot Ulcer. J Diabetes Res 2017; 2017:9328347. [PMID: 28386568 PMCID: PMC5366201 DOI: 10.1155/2017/9328347] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 02/28/2017] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs), an ideal cell source for regenerative therapy with no ethical issues, play an important role in diabetic foot ulcer (DFU). Growing evidence has demonstrated that MSCs transplantation can accelerate wound closure, ameliorate clinical parameters, and avoid amputation. In this review, we clarify the mechanism of preclinical studies, as well as safety and efficacy of clinical trials in the treatment of DFU. Bone marrow-derived mesenchymal stem cells (BM-MSCs), compared with MSCs derived from other tissues, may be a suitable cell type that can provide easy, effective, and cost-efficient transplantation to treat DFU and protect patients from amputation.
Collapse
Affiliation(s)
- Yue Cao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Chenglin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
- *Guixia Wang:
| |
Collapse
|
21
|
van Dongen JA, Stevens HP, Parvizi M, van der Lei B, Harmsen MC. The fractionation of adipose tissue procedure to obtain stromal vascular fractions for regenerative purposes. Wound Repair Regen 2016; 24:994-1003. [PMID: 27717133 DOI: 10.1111/wrr.12482] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/03/2016] [Indexed: 12/27/2022]
Abstract
Autologous adipose tissue transplantation is clinically used to reduce dermal scarring and to restore volume loss. The therapeutic benefit on tissue damage more likely depends on the stromal vascular fraction of adipose tissue than on the adipocyte fraction. This stromal vascular fraction can be obtained by dissociation of adipose tissue, either enzymatically or mechanical. Enzymatic dissociation procedures are time-consuming and expensive. Therefore, we developed a new inexpensive mechanical dissociation procedure to obtain the stromal vascular fraction from adipose tissue in a time sparing way, which is directly available for therapeutic injection. This mechanical dissociation procedure is denoted as the fractionation of adipose tissue (FAT) procedure. The FAT procedure was performed in eleven patients. The composition of the FAT-stromal vascular fraction was characterized by immunohistochemistry. Adipose derived stromal cells isolated from the FAT-stromal vascular fraction were compared with adipose derived stromal cells isolated from nondissociated adipose tissue (control) for their CD-surface marker expression, differentiation and colony forming unit capacity. Case reports demonstrated the therapeutic effect of the FAT-stromal vascular fraction. The FAT-stromal vascular fraction is an enrichment of extracellular matrix containing a microvasculature and culturable adipose derived stromal cells. Adipose derived stromal cells isolated from FAT-stromal vascular fraction did not differ from adipose derived stromal cells isolated from the control group in CD-surface marker expression, differentiation and colony forming unit capacity. The FAT procedure is a rapid effective mechanical dissociation procedure to generate FAT-stromal vascular fraction ready for injection with all its therapeutic components of adipose tissue: it contains culturable adipose derived stromal cells embedded in their natural supportive extracellular matrix together with the microvasculature.
Collapse
Affiliation(s)
- Joris A van Dongen
- Plastic Surgery Department, Bergman Clinics, Rijswijk, The Netherlands.,Department of Pathology & Medical Biology, University of Groningen and University Medical Centre of Groningen, Groningen, The Netherlands.,Department of Plastic Surgery, University of Groningen and University Medical Centre of Groningen, Groningen, The Netherlands
| | | | - Mojtaba Parvizi
- Department of Pathology & Medical Biology, University of Groningen and University Medical Centre of Groningen, Groningen, The Netherlands
| | - Berend van der Lei
- Department of Plastic Surgery, University of Groningen and University Medical Centre of Groningen, Groningen, The Netherlands.,Plastic Surgery Department, Bergman Clinics, Heerenveen and Zwolle, The Netherlands
| | - Martin C Harmsen
- Department of Pathology & Medical Biology, University of Groningen and University Medical Centre of Groningen, Groningen, The Netherlands
| |
Collapse
|
22
|
Bi Y, Han X, Zhong H, Xu K. Correspondence on: "Therapeutic Prospect of Adipose-Derived Stromal Cells for Treatment of Abdominal Aortic Aneurysm". Stem Cells Dev 2015; 24:1831-2. [PMID: 25945966 DOI: 10.1089/scd.2015.0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Yonghua Bi
- 1 Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, China .,2 Department of Radiology, The First Affiliated Hospital of China Medical University , Shenyang, China
| | - Xinwei Han
- 1 Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, China
| | - Hongshan Zhong
- 2 Department of Radiology, The First Affiliated Hospital of China Medical University , Shenyang, China
| | - Ke Xu
- 2 Department of Radiology, The First Affiliated Hospital of China Medical University , Shenyang, China
| |
Collapse
|