1
|
Bian C, Chen G, Cheng X, Gu H, Huang Z, Zhou K. Facile fabrication of nano-bioactive glass functionalized blended hydrogel with nucleus pulposus-derived MSCs to improve regeneration potential in treatment of disc degeneration by in vivo rat model. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2025; 63:102790. [PMID: 39414222 DOI: 10.1016/j.nano.2024.102790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Orthopaedic medicine often treats intervertebral disc degeneration (IVDD), which is caused by nucleus pulposus (NP) tissue damage and mechanical stress. Bioactive glasses (BGs), widely used for bone regeneration, can incorporate therapeutic ions into their network. Manganese (Mn) activates human osteoblast integrins, proliferation, and spreading. The CMnBGNPs-NPMSCs are carboxymethyl cellulose hydrogels functionalized with MnBGsNPs and NP-derived mesenchymal stem cells to treat IVDD. To ensure stability and biocompatibility of CMnBGNPs-NPMSCs were characterized for rheological properties like gelation time and swelling ratio. Gene expression analysis of PAX1, FOXF1, CA12, HBB, and OVOS2 via qRT-PCR further assessed the hydrogel's characteristics. Rat models with induced IVDD had hydrogel-MSC composite injected into their intervertebral discs for in vivo studies. Histological examination, immunohistochemical staining for inflammation and disc regeneration markers, and disc height assessments assessed therapeutic efficacy. CMnBGNPs-NPMSCs show promising results for IVDD treatment, offering a novel therapeutic strategy with clinical implications for degenerative disc diseases.
Collapse
Affiliation(s)
- Chong Bian
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Guangnan Chen
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Xiangyang Cheng
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Huijie Gu
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Zhongyue Huang
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Kaifeng Zhou
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai 201199, China.
| |
Collapse
|
2
|
Arriola-Alvarez I, Jaunarena I, Izeta A, Lafuente H. Progenitor Cell Sources for 3D Bioprinting of Lymphatic Vessels and Potential Clinical Application. Tissue Eng Part A 2024; 30:353-366. [PMID: 37950710 DOI: 10.1089/ten.tea.2023.0204] [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: 11/13/2023] Open
Abstract
The lymphatic system maintains tissue fluid homeostasis and it is involved in the transport of nutrients and immunosurveillance. It also plays a pivotal role in both pathological and regenerative processes. Lymphatic development in the embryo occurs by polarization and proliferation of lymphatic endothelial cells from the lymph sacs, that is, lymphangiogenesis. Alternatively, lymphvasculogenesis further contributes to the formation of lymphatic vessels. In adult tissues, lymphatic formation rarely occurs under physiological conditions, being restricted to pathological processes. In lymphvasculogenesis, progenitor cells seem to be a source of lymphatic vessels. Indeed, mesenchymal stem cells, adipose stem cells, endothelial progenitor cells, and colony-forming endothelial cells are able to promote lymphatic regeneration by different mechanisms, such as direct differentiation and paracrine effects. In this review, we summarize what is known on the diverse stem/progenitor cell niches available for the lymphatic system, emphasizing the potential that these cells hold for lymphatic tissue engineering through 3D bioprinting and their translation to clinical application.
Collapse
Affiliation(s)
- Inazio Arriola-Alvarez
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| | - Ibon Jaunarena
- Gynecology Oncology Unit, Donostia University Hospital, Donostia-San Sebastián, Spain
- Obstetrics and Gynaecology Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- University of the Basque Country (UPV/EHU), Department of Medical Surgical Specialties, Leioa, Spain
| | - Ander Izeta
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- Department of Biomedical Engineering and Sciences, Tecnun-University of Navarra, Donostia-San Sebastián, Spain
| | - Héctor Lafuente
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| |
Collapse
|
3
|
Asano Y, Shimoda H, Okano D, Matsusaki M, Akashi M. Lymphatic Drainage-Promoting Effects by Engraftment of Artificial Lymphatic Vascular Tissue Based on Human Adipose Tissue-Derived Mesenchymal Stromal Cells in Mice. J Tissue Eng Regen Med 2023; 2023:7626767. [PMID: 40226423 PMCID: PMC11919049 DOI: 10.1155/2023/7626767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/09/2023] [Accepted: 09/28/2023] [Indexed: 04/15/2025]
Abstract
Regenerative medicine using lymphatic vascular engineering is a promising approach for treating lymphedema. However, its development lags behind that of artificial blood vascular tissue for ischemic diseases. In this study, we constructed artificial 3D lymphatic vascular tissue, termed ASCLT, by co-cultivation of ECM-nanofilm-coated human adipose tissue-derived mesenchymal stromal cells (hASCs) and human dermal lymphatic endothelial cells (HDLECs). The effect of hASCs in lymphatic vessel network formation was evaluated by comparison with the tissue based on fibroblasts, termed FbLT. Our results showed that the density of lymphatic vascular network in ASCLT was higher than that in FbLT, demonstrating a promoting effect of hASCs on lymphatic vascular formation. This result was also supported by higher levels of lymphangiogenesis-promoting factors, such as bFGF, HGF, and VEGF-A in ASCLT than in FbLT. To evaluate the therapeutic effects, FbLTs and ASCLTs were subcutaneously transplanted to mouse hindlimb lymphatic drainage interruption models by removal of popliteal and subiliac lymph nodes. Despite the restricted engraftment of lymphatic vessels, ASCLT promoted regeneration of irregular and diverse lymphatic drainage in the skin, as visualized by indocyanine green imaging. Moreover, transplantation of ASCLT to the popliteal lymph node resection area also resulted in lymphatic drainage regeneration. Histological analysis of the generated drainage visualized by FITC-dextran injection revealed that the drainage was localized in the subcutaneous area shallower than the dermal muscle. These findings demonstrate that ASCLT promotes lymphatic drainage in vivo and that hASCs can serve as an autologous source for treatment of secondary lymphedema by tissue engineering.
Collapse
Affiliation(s)
- Yoshiya Asano
- Department of Neuroanatomy, Cell Biology and Histology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Hiroshi Shimoda
- Department of Neuroanatomy, Cell Biology and Histology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
- Department of Anatomical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Daisuke Okano
- Department of Neuroanatomy, Cell Biology and Histology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Mitsuru Akashi
- Building Block Science, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamada-oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
4
|
Saha A, Kolonin MG, DiGiovanni J. Obesity and prostate cancer - microenvironmental roles of adipose tissue. Nat Rev Urol 2023; 20:579-596. [PMID: 37198266 DOI: 10.1038/s41585-023-00764-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 05/19/2023]
Abstract
Obesity is known to have important roles in driving prostate cancer aggressiveness and increased mortality. Multiple mechanisms have been postulated for these clinical observations, including effects of diet and lifestyle, systemic changes in energy balance and hormonal regulation and activation of signalling by growth factors and cytokines and other components of the immune system. Over the past decade, research on obesity has shifted towards investigating the role of peri-prostatic white adipose tissue as an important source of locally produced factors that stimulate prostate cancer progression. Cells that comprise white adipose tissue, the adipocytes and their progenitor adipose stromal cells (ASCs), which proliferate to accommodate white adipose tissue expansion in obesity, have been identified as important drivers of obesity-associated cancer progression. Accumulating evidence suggests that adipocytes are a source of lipids that are used by adjacent prostate cancer cells. However, results of preclinical studies indicate that ASCs promote tumour growth by remodelling extracellular matrix and supporting neovascularization, contributing to the recruitment of immunosuppressive cells, and inducing epithelial-mesenchymal transition through paracrine signalling. Because epithelial-mesenchymal transition is associated with cancer chemotherapy resistance and metastasis, ASCs are considered to be potential targets of therapies that could be developed to suppress cancer aggressiveness in patients with obesity.
Collapse
Affiliation(s)
- Achinto Saha
- Division of Pharmacology and Toxicology and Dell Paediatric Research Institute, The University of Texas at Austin, Austin, TX, USA
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Disease, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA.
| | - John DiGiovanni
- Division of Pharmacology and Toxicology and Dell Paediatric Research Institute, The University of Texas at Austin, Austin, TX, USA.
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, USA.
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
5
|
Moradi-Gharibvand N, Hashemibeni B. The Effect of Stem Cells and Vascular Endothelial Growth Factor on Cancer Angiogenesis. Adv Biomed Res 2023; 12:124. [PMID: 37434939 PMCID: PMC10331557 DOI: 10.4103/abr.abr_378_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/17/2022] [Accepted: 04/24/2022] [Indexed: 07/13/2023] Open
Abstract
The formation of new vessels from pre-existing vessels is known as angiogenesis. The process is controlled by stimuli and inhibitors. Angiogenesis starts as a result of the unbalance of these factors, where balance has a tendency toward the stimulus. One of the most important factors promoting angiogenesis is the vascular endothelial growth factor (VEGF). In addition to being involved in vascular regeneration in normal tissues, VEGF also takes part in tumor tissue angiogenesis. These factors affect endothelial cells (ECs) directly as well as differentiate tumor cells from endothelial cells and play an active role in tumor tissue angiogenesis. Angiogenesis partakes in the growth and proliferation of tumor tissue. Because anti-angiogenic treatment is favorable in existing cancer therapies, the potential benefits should be considered. One of these new therapies is cell therapy using mesenchymal stem cells (MSCs). Research on MSCs remains controversial because much of the earlier research on MSCs has shown their effectiveness, but more recent research has identified harmful effects of these cells. This article reviews the role of stem cells and their secretions in the angiogenesis of tumor tissues.
Collapse
Affiliation(s)
- Nahid Moradi-Gharibvand
- Abadan University of Medical Sciences, Abadan, Iran
- Department of Anatomical Sciences and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
6
|
A Novel Dressing Composed of Adipose Stem Cells and Decellularized Wharton's Jelly Facilitated Wound Healing and Relieved Lymphedema by Enhancing Angiogenesis and Lymphangiogenesis in a Rat Model. J Funct Biomater 2023; 14:jfb14020104. [PMID: 36826903 PMCID: PMC9960849 DOI: 10.3390/jfb14020104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Lymphedema causes tissue swelling due to the accumulation of lymphatic fluid in the tissue, which delays the process of wound-healing. Developing effective treatment options of lymphedema is still an urgent issue. In this study, we aim to fabricate tissue-engineered moist wound dressings with adipose stem cells (ASCs) and decellularized Wharton's jelly (dWJ) from the human umbilical cord in order to ameliorate lymphedema. Rat ASCs were proliferated and an apparent layer was observed on dWJ at day 7 and 14. A rat tail lymphedema model was developed to evaluate the efficacy of the treatment. Approximately 1 cm of skin near the base of the rat tail was circularly excised. The wounds were treated by secondary healing (control) (n = 5), decellularized Wharton's jelly (n = 5) and ASC-seeded dWJ (n = 5). The wound-healing rate and the tail volume were recorded once a week from week one to week five. Angiogenesis and lymphangiogenesis were assessed by immunochemistry staining with anti-CD31 and anti-LYVE1. The results showed that the wound-healing rate was faster and the tail volume was lesser in the ASC-seeded dWJ group than in the control group. More CD31+ and LYVE-1+ cells were observed at the wound-healing area in the ASC-seeded dWJ group than in the control group. This proves that tissue-engineered moist wound dressings can accelerate wound-healing and reduce lymphedema by promoting angiogenesis and lymphangiogenesis.
Collapse
|
7
|
Nagahara AI, Homma J, Ryu B, Sekine H, Higashi Y, Shimizu T, Kawamata T. Networked lymphatic endothelial cells in a transplanted cell sheet contribute to form functional lymphatic vessels. Sci Rep 2022; 12:21698. [PMID: 36522421 PMCID: PMC9755306 DOI: 10.1038/s41598-022-26041-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
This study evaluated whether cell sheets containing a network of lymphatic endothelial cells (LECs) promoted lymphangiogenesis after transplantation in vivo. Cell sheets with a LEC network were constructed by co-culturing LECs and adipose-derived stem cells (ASCs) on temperature-responsive culture dishes. A cell ratio of 3:2 (vs. 1:4) generated networks with more branches and longer branch lengths. LEC-derived lymphatic vessels were observed 2 weeks after transplantation of a three-layered cell sheet construct onto rat gluteal muscle. Lymphatic vessel number, diameter and depth were greatest for a construct comprising two ASC sheets stacked on a LEC/ASC (3:2 ratio) sheet. Transplantation of this construct in a rat model of femoral lymphangiectomy led to the formation of functional lymphatic vessels containing both transplanted and host LECs. Further development of this technique may lead to a new method of promoting lymphangiogenesis.
Collapse
Affiliation(s)
- Ayumi Inoue Nagahara
- grid.410818.40000 0001 0720 6587Department of Neurosurgery, Graduate School of Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Jun Homma
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Bikei Ryu
- grid.488555.10000 0004 1771 2637Department of Neurosurgery, Tokyo Women’s Medical University Hospital, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Hidekazu Sekine
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Yuhei Higashi
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan ,Tokaihit Co., Ltd., Shizuoka, Japan
| | - Tatsuya Shimizu
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Takakazu Kawamata
- grid.488555.10000 0004 1771 2637Department of Neurosurgery, Tokyo Women’s Medical University Hospital, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| |
Collapse
|
8
|
Saha A, Hamilton-Reeves J, DiGiovanni J. White adipose tissue-derived factors and prostate cancer progression: mechanisms and targets for interventions. Cancer Metastasis Rev 2022; 41:649-671. [PMID: 35927363 PMCID: PMC9474694 DOI: 10.1007/s10555-022-10056-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/27/2022] [Indexed: 12/01/2022]
Abstract
Obesity represents an important risk factor for prostate cancer, driving more aggressive disease, chemoresistance, and increased mortality. White adipose tissue (WAT) overgrowth in obesity is central to the mechanisms that lead to these clinical observations. Adipose stromal cells (ASCs), the progenitors to mature adipocytes and other cell types in WAT, play a vital role in driving PCa aggressiveness. ASCs produce numerous factors, especially chemokines, including the chemokine CXCL12, which is involved in driving EMT and chemoresistance in PCa. A greater understanding of the impact of WAT in obesity-induced progression of PCa and the underlying mechanisms has begun to provide opportunities for developing interventional strategies for preventing or offsetting these critical events. These include weight loss regimens, therapeutic targeting of ASCs, use of calorie restriction mimetic compounds, and combinations of compounds as well as specific receptor targeting strategies.
Collapse
Affiliation(s)
- Achinto Saha
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78723, USA
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, 78723, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA
| | - Jill Hamilton-Reeves
- Departments of Urology and Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78723, USA.
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, 78723, USA.
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA.
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA.
| |
Collapse
|
9
|
Jia W, He W, Wang G, Goldman J, Zhao F. Enhancement of Lymphangiogenesis by Human Mesenchymal Stem Cell Sheet. Adv Healthc Mater 2022; 11:e2200464. [PMID: 35678079 PMCID: PMC11932734 DOI: 10.1002/adhm.202200464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/08/2022] [Indexed: 01/24/2023]
Abstract
Preparation of human mesenchymal stem cell (hMSC) suspension for lymphedema treatment relies on conventional enzymatic digestion methods, which severely disrupts cell-cell and cell-extracellular matrix (ECM) connections, and drastically impairs cell retention and engraftment after transplantation. The objective of the present study is to evaluate the ability of hMSC-secreted ECM to augment lymphangiogenesis by using an in vitro coculturing model of hMSC sheets with lymphatic endothelial cells (LECs) and an in vivo mouse tail lymphedema model. Results demonstrate that the hMSC-secreted ECM augments the formation of lymphatic capillary-like structure by a factor of 1.2-3.6 relative to the hMSC control group, by serving as a prolymphangiogenic growth factor reservoir and facilitating cell regenerative activities. hMSC-derived ECM enhances MMP-2 mediated matrix remodeling, increases the synthesis of collagen IV and laminin, and promotes lymphatic microvessel-like structure formation. The injection of rat MSC sheet fragments into a mouse tail lymphedema model confirms the benefits of the hMSC-derived ECM by stimulating lymphangiogenesis and wound closure.
Collapse
Affiliation(s)
- Wenkai Jia
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, Emerging Technologies Building, College Station, TX, 77843, USA
| | - Weilue He
- Department of Biomedical Engineering, Michigan Technological University, Minerals & Materials Building, 1400 Townsend Drive, Room 309, Houghton, MI, 44931, USA
| | - Guifang Wang
- Department of Biomedical Engineering, Michigan Technological University, Minerals & Materials Building, 1400 Townsend Drive, Room 309, Houghton, MI, 44931, USA
| | - Jeremy Goldman
- Department of Biomedical Engineering, Michigan Technological University, Minerals & Materials Building, 1400 Townsend Drive, Room 309, Houghton, MI, 44931, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, Emerging Technologies Building, College Station, TX, 77843, USA
| |
Collapse
|
10
|
Hromada C, Hartmann J, Oesterreicher J, Stoiber A, Daerr A, Schädl B, Priglinger E, Teuschl-Woller AH, Holnthoner W, Heinzel J, Hercher D. Occurrence of Lymphangiogenesis in Peripheral Nerve Autografts Contrasts Schwann Cell-Induced Apoptosis of Lymphatic Endothelial Cells In Vitro. Biomolecules 2022; 12:820. [PMID: 35740945 PMCID: PMC9221261 DOI: 10.3390/biom12060820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/04/2023] Open
Abstract
Peripheral nerve injuries pose a major clinical concern world-wide, and functional recovery after segmental peripheral nerve injury is often unsatisfactory, even in cases of autografting. Although it is well established that angiogenesis plays a pivotal role during nerve regeneration, the influence of lymphangiogenesis is strongly under-investigated. In this study, we analyzed the presence of lymphatic vasculature in healthy and regenerated murine peripheral nerves, revealing that nerve autografts contained increased numbers of lymphatic vessels after segmental damage. This led us to elucidate the interaction between lymphatic endothelial cells (LECs) and Schwann cells (SCs) in vitro. We show that SC and LEC secretomes did not influence the respective other cell types' migration and proliferation in 2D scratch assay experiments. Furthermore, we successfully created lymphatic microvascular structures in SC-embedded 3D fibrin hydrogels, in the presence of supporting cells; whereas SCs seemed to exert anti-lymphangiogenic effects when cultured with LECs alone. Here, we describe, for the first time, increased lymphangiogenesis after peripheral nerve injury and repair. Furthermore, our findings indicate a potential lymph-repellent property of SCs, thereby providing a possible explanation for the lack of lymphatic vessels in the healthy endoneurium. Our results highlight the importance of elucidating the molecular mechanisms of SC-LEC interaction.
Collapse
Affiliation(s)
- Carina Hromada
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria; (C.H.); (A.D.); (A.H.T.-W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
| | - Jaana Hartmann
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Johannes Oesterreicher
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Anton Stoiber
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Anna Daerr
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria; (C.H.); (A.D.); (A.H.T.-W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
| | - Barbara Schädl
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
- University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Eleni Priglinger
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Andreas H. Teuschl-Woller
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria; (C.H.); (A.D.); (A.H.T.-W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
| | - Wolfgang Holnthoner
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Johannes Heinzel
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Unfallklinik Tuebingen, University of Tuebingen, 72076 Tuebingen, Germany
| | - David Hercher
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| |
Collapse
|
11
|
Qi S, Ye L, Hu L, Pan J. In Vitro Induction of Human Dental Pulp Stem Cells to Lymphatic Endothelial Cells. Cell Reprogram 2022; 24:186-194. [PMID: 35559757 DOI: 10.1089/cell.2021.0106] [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: 02/05/2023] Open
Abstract
Lymphedema is a progressive and irreversible disease due to the lymphatic system disorder. Conservative and surgical therapies are either ineffective or impractical. Currently, mesenchymal stem cells (MSCs)-based therapies seem to be the most promising treatment for lymphedema. The MSCs promote lymphangiogenesis through the paracrine approach or by directly differentiating into lymphatic endothelial cells (LECs) under the induction of growth factors. Human dental pulp stem cells (hDPSCs) have been suggested to play important roles in tissue regeneration, making it an attractive candidate for the lymphedema treatment. In this study, to evaluate the potential role of hDPSCs in the clinical application for lymphedema treatment, we induced the hDPSCs with vascular endothelial growth factor-C (VEGF-C) and investigated the lymphangiogenic differentiation potential of hDPSCs in vitro. We found that under the VEGF-C induction, hDPSCs demonstrated upregulated LECs specific markers, promoted cell proliferation and migration, and increased tube formation, all of which contributed to their differentiation into LECs in vitro.
Collapse
Affiliation(s)
- Shuqun Qi
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, Sichuan University, West China Hospital of Stomatology, Chengdu, China
| | - Li Ye
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, Sichuan University, West China Hospital of Stomatology, Chengdu, China
| | - Liru Hu
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, Sichuan University, West China Hospital of Stomatology, Chengdu, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, Sichuan University, West China Hospital of Stomatology, Chengdu, China
| |
Collapse
|
12
|
Dong D, Wang H, Chen L, Wang W, Liu T. Hormone Therapy: A Potential Risk Factor Affecting Survival and Functional Restoration of Transplanted Lymph Nodes. Front Pharmacol 2022; 13:853859. [PMID: 35431925 PMCID: PMC9008310 DOI: 10.3389/fphar.2022.853859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/09/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Transplantation of lymph nodes (LNs) is an increasingly popular option for treating lymphedema. Increasing evidence indicates an intrinsic correlation between estrogen signaling and the lymphatic system. We explored the effects of 17β estradiol and antiestrogen treatment (tamoxifen) on the survival and functional restoration of transplanted popliteal lymph nodes (PLNs). Methods: A total of forty-eight ovariectomized mice were divided into three groups of 16: OVX + E2 (treated with 17β-estradiol), OVX + TMX (treated with tamoxifen), and OVX (control; treated with olive oil as a solvent). After 2 weeks, PLNs were transplanted. Then, reconnections of lymphatic vessels were observed, and the morphology and survival of transplanted PLNs were evaluated 4 weeks after transplantation. T cells, B cells, lymphatic vessels, and high endothelial venules (HEVs) were subjected to immunofluorescence staining or immunohistochemical staining and quantified. Results: The percentage of lymphatic reconnections was 93.75% in the OVX + E2 group, 68.75% in the OVX + TMX group, and 75% in the OVX group. Surviving PLNs were observed in 16 of 16 in the OVX + E2 group, seven of 16 in the OVX + TMX group, and 13 of 16 in the OVX group. The mean size of PLNs in the largest cross section of the OVX + TMX group was significantly lower than that in the other groups. The distributions of B cells and T cells in surviving PLNs were similar to those in normal LNs. The ratio of dilated HEVs/total HEVs and density of lymphatic vessels in the OVX + E2 group were the highest among the three groups, whereas the lowest ratio and density were observed in the OVX + TMX group. Conclusion: Tamoxifen treatment might lead to cellular loss of transplanted LNs and interfere with the structural reconstruction and functional restoration, thereby inhibiting the survival of transplanted PLNs. Estrogen treatment facilitated the maintenance and regeneration of functional HEVs as well as lymphangiogenesis.
Collapse
Affiliation(s)
- Dong Dong
- Department of Plastic and Aesthetic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Heng Wang
- Department of Plastic and Aesthetic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Liang Chen
- Department of Plastic and Aesthetic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Wei Wang
- Department of Plastic and Aesthetic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Tianyi Liu
- Department of Plastic and Aesthetic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- *Correspondence: Tianyi Liu,
| |
Collapse
|
13
|
Yang Y, Li XB, Li Y, Li TX, Li P, Deng GM, Guo Q, Zhou X, Chen XH. Extracellular Vesicles Derived From Hypoxia-Conditioned Adipose-Derived Mesenchymal Stem Cells Enhance Lymphangiogenesis. Cell Transplant 2022; 31:9636897221107536. [PMID: 35861534 PMCID: PMC9310282 DOI: 10.1177/09636897221107536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Extracellular vesicles from adipose-derived mesenchymal stem cells (ADSCs) play an important role in lymphangiogenesis; however, the underlying mechanisms are not fully understood. In this study, we aimed to investigate the function of extracellular vesicles secreted by hypoxia-conditioned ADSCs in lymphangiogenesis and explore the potential molecular mechanisms. Extracellular vesicles were extracted from ADSCs cultured under hypoxia or normoxia conditions. The uptake of extracellular vesicles by lymphatic endothelial cells (LECs) was detected by immunofluorescence staining. The effects of extracellular vesicles on the viability, migration, and tube formation of LECs were determined by CCK-8 assay, migration assay, and tube formation assay, respectively. Molecules and pathway involved in lymphangiogenesis mediated by ADSC-derived extracellular vesicles were analyzed by luciferase reporter assay, qRT-polymerase chain reaction (PCR), and Western blot. Hypoxia ADSC-derived extracellular vesicles (H-ADSC/evs) significantly enhanced the proliferation, migration, and tube formation of LECs. Hypoxia decreased the expression of miR-129 in ADSC-derived extracellular vesicles. Overexpression of miR-129 counteracted the promoting effect of H-ADSC/evs on lymphangiogenesis. In addition, decreased exosomal miR-129 expression resulted in upregulation of HMGB1 in LECs, which led to AKT activation and lymphangiogenesis enhancement. Our data reveal that extracellular vesicles derived from hypoxia-conditioned ADSCs induce lymphangiogenesis, and this effect is mediated by miR-129/HMGB1/AKT signaling. Our findings imply that hypoxia ADSC-isolated extracellular vesicles may represent as a valuable target for the treatment of diseases associated with lymphatic remodeling.
Collapse
Affiliation(s)
- Yi Yang
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xu-Bo Li
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Li
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tian-Xiao Li
- Department of Pharmacy, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ping Li
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guang-Mao Deng
- Department of Orthopedic, Huiya Hospital, The First Affiliated Hospital of Sun Yat-sen University, Huizhou, China
| | - Qiang Guo
- Department of Orthopedic, Huiya Hospital, The First Affiliated Hospital of Sun Yat-sen University, Huizhou, China
| | - Xiang Zhou
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiao-Hu Chen
- Department of Orthopedic, Huiya Hospital, The First Affiliated Hospital of Sun Yat-sen University, Huizhou, China
| |
Collapse
|
14
|
Molecular mechanisms of cyclic phosphatidic acid-induced lymphangiogenic actions in vitro. Microvasc Res 2021; 139:104273. [PMID: 34699844 DOI: 10.1016/j.mvr.2021.104273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 11/21/2022]
Abstract
The lymphatic system plays important roles in various physiological and pathological phenomena. As a bioactive phospholipid, lysophosphatidic acid (LPA) has been reported to function as a lymphangiogenic factor as well as some growth factors, yet the involvement of phospholipids including LPA and its derivatives in lymphangiogenesis is not fully understood. In the present study, we have developed an in-vitro lymphangiogenesis model (termed a collagen sandwich model) by utilizing type-I collagen, which exists around the lymphatic endothelial cells of lymphatic capillaries in vivo. The collagen sandwich model has revealed that cyclic phosphatidic acid (cPA), and not LPA, augmented the tube formation of human dermal lymphatic endothelial cells (HDLECs). Both cPA and LPA increased the migration of HDLECs cultured on the collagen. As the gene expression of LPA receptor 6 (LPA6) was predominantly expressed in HDLECs, a siRNA experiment against LPA6 attenuated the cPA-mediated tube formation. A synthetic LPA1/3 inhibitor, Ki16425, suppressed the cPA-augmented tube formation and migration of the HDLECs, and the LPA-induced migration. The activity of Rho-associated protein kinase (ROCK) located at the downstream of the LPA receptors was augmented in both the cPA- and LPA-treated cells. A potent ROCK inhibitor, Y-27632, suppressed the cPA-dependent tube formation but not the migration of the HDLECs. Furthermore, cPA, but not LPA, augmented the gene expression of VE-cadherin and β-catenin in the HDLECs. These results provide novel evidence that cPA facilitates the capillary-like morphogenesis and the migration of HDLECs through LPA6/ROCK and LPA1/3 signaling pathways in concomitance with the augmentation of VE-cadherin and β-catenin expression. Thus, cPA is likely to be a potent lymphangiogenic factor for the initial lymphatics adjacent to type I collagen under physiological conditions.
Collapse
|
15
|
Use of adipose-derived stem cells in lymphatic tissue engineering and regeneration. Arch Plast Surg 2021; 48:559-567. [PMID: 34583446 PMCID: PMC8490113 DOI: 10.5999/aps.2021.00339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/16/2021] [Indexed: 12/11/2022] Open
Abstract
The potential to differentiate into different cell lines, added to the easy and cost-effective method of extraction, makes adipose-derived stem cells (ADSCs) an object of interest in lymphedema treatment. Our study’s goal was to conduct a comprehensive systematic review of the use of ADSCs in lymphatic tissue engineering and regeneration. On July 23, 2019, using PubMed/MEDLINE, Cochrane Clinical Answers, Cochrane Central Register of Controlled Trials, and Embase databases, we conducted a systematic review of published literature on the use of ADSCs in lymphatic tissue engineering and regeneration. There were no language or time frame limitations, and the following search strategy was applied: ((Adipose stem cell) OR Adipose-derived stem cell)) AND ((Lymphedema) OR Breast Cancer Lymphedema). Only original research manuscripts were included. Fourteen studies fulfilled the inclusion criteria. Eleven studies were experimental (in vitro or in vivo in animals), and only three were clinical. Publications on the topic demonstrated that ADSCs promote lymphangiogenesis, and its effect could be enhanced by modulation of vascular endothelial growth factor-C, interleukin-7, prospero homeobox protein 1, and transforming growth factor-β1. Pilot clinical studies included 11 patients with breast cancer-related lymphedema, and no significant side effects were present at 12-month follow-up. Literature on the use of ADSCs in lymphatic tissue engineering and regeneration demonstrated promising data. Clinical evidence is still in its infancy, but the scientific community agrees that ADSCs can be useful in regenerative lymphangiogenesis. Data collected in this review indicate that unprecedented advances in lymphedema treatment can be anticipated in the upcoming years.
Collapse
|
16
|
Singla B, Lin HP, Chen A, Ahn W, Ghoshal P, Cherian-Shaw M, White J, Stansfield BK, Csányi G. Role of R-spondin 2 in arterial lymphangiogenesis and atherosclerosis. Cardiovasc Res 2021; 117:1489-1509. [PMID: 32750106 PMCID: PMC8152716 DOI: 10.1093/cvr/cvaa244] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/16/2020] [Accepted: 07/30/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS Impaired lymphatic drainage of the arterial wall results in intimal lipid accumulation and atherosclerosis. However, the mechanisms regulating lymphangiogenesis in atherosclerotic arteries are not well understood. Our studies identified elevated levels of matrix protein R-spondin 2 (RSPO2) in atherosclerotic arteries. In this study, we investigated the role of RSPO2 in lymphangiogenesis, arterial cholesterol efflux into lesion-draining lymph nodes (LNs) and development of atherosclerosis. METHODS AND RESULTS The effect of RSPO2 on lymphangiogenesis was investigated using human lymphatic endothelial cells (LEC) in vitro and implanted Matrigel plugs in vivo. Cellular and molecular approaches, pharmacological agents, and siRNA silencing of RSPO2 receptor LGR4 were used to investigate RSPO2-mediated signalling in LEC. In vivo low-density lipoprotein (LDL) tracking and perivascular blockade of RSPO2-LGR4 signalling using LGR4-extracellular domain (ECD) pluronic gel in hypercholesterolemic mice were utilized to investigate the role of RSPO2 in arterial reverse cholesterol transport and atherosclerosis. Immunoblotting and imaging experiments demonstrated increased RSPO2 expression in human and mouse atherosclerotic arteries compared to non-atherosclerotic controls. RSPO2 treatment inhibited lymphangiogenesis both in vitro and in vivo. LGR4 silencing and inhibition of RSPO2-LGR4 signalling abrogated RSPO2-induced inhibition of lymphangiogenesis. Mechanistically, we found that RSPO2 suppresses PI3K-AKT-endothelial nitric oxide synthase (eNOS) signalling via LGR4 and inhibits activation of the canonical Wnt-β-catenin pathway. ApoE-/- mice treated with LGR4-ECD developed significantly less atherosclerosis compared with control treatment. Finally, increased arterial lymphatic vessel density and improved lymphatic drainage of fluorescently labelled LDL to deep cervical LNs were observed in LGR4-ECD-treated mice. CONCLUSION These findings demonstrate that RSPO2 inhibits lymphangiogenesis via LGR4 and downstream impairment of AKT-eNOS-nitric oxide signalling. These results may also inform new therapeutic strategies to promote lymphangiogenesis and improve cholesterol efflux from atherosclerotic arteries.
Collapse
Affiliation(s)
- Bhupesh Singla
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
| | - Hui-Ping Lin
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
| | - Alex Chen
- Medical Scholars Program, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
| | - WonMo Ahn
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
| | - Pushpankur Ghoshal
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
| | - Joseph White
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, BF 104, Augusta, GA 30912, USA
| | - Brian K Stansfield
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
- Department of Pediatrics, Medical College of Georgia at Augusta University, 1120 15th Street, BI6031, Augusta, GA 30912, USA
| | - Gábor Csányi
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
| |
Collapse
|
17
|
Zhang L, Ocansey DKW, Liu L, Olovo CV, Zhang X, Qian H, Xu W, Mao F. Implications of lymphatic alterations in the pathogenesis and treatment of inflammatory bowel disease. Biomed Pharmacother 2021; 140:111752. [PMID: 34044275 DOI: 10.1016/j.biopha.2021.111752] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/06/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by intense immune dysregulation, gut microbiota imbalance, and intestinal epithelium destruction. Among the factors that contribute to the pathogenesis of IBD, lymphatics have received less attention, hence less studied, characterized, and explored. However, in recent years, the role of the lymphatic system in gastrointestinal pathophysiology continues to be highlighted. This paper examines the implications of lymphatic changes in IBD pathogenesis related to immune cells, gut microbiota, intestinal and mesenteric epithelial barrier integrity, and progression to colorectal cancer (CRC). Therapeutic targets of lymphatics in IBD studies are also presented. Available studies indicate that lymph nodes and other secondary lymphatic tissues, provide highly specialized microenvironments for mounting effective immune responses and that lymphatic integrity plays a significant role in small intestine homeostasis, where the lymphatic vasculature effectively controls tissue edema, leukocyte exit, bacterial antigen, and inflammatory chemokine clearance. In IBD, there are functional and morphological alterations in intestinal and mesenteric lymphatic vessels (more profoundly in Crohn's disease [CD] compared to ulcerative colitis [UC]), including lymphangiogenesis, lymphangiectasia, lymphadenopathy, and lymphatic vasculature blockade, affecting not only immunity but gut microbiota and epithelial barrier integrity. While increased lymphangiogenesis is primarily associated with a good prognosis of IBD, increased lymphangiectasia, lymphadenopathy, and lymphatic vessel occlusion correlate with poor prognosis. IBD therapies that target the lymphatic system seek to increase lymphangiogenesis via induction of lymphangiogenic factors and inhibition of its antagonists. The resultant increased lymphatic flow coupled with other anti-inflammatory activities restores gut homeostasis.
Collapse
Affiliation(s)
- Lu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China; Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Lianqin Liu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Chinasa Valerie Olovo
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China; Department of Microbiology, University of Nigeria, Nsukka 410001, Nigeria
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Hui Qian
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Wenrong Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China.
| |
Collapse
|
18
|
Jia W, Hitchcock-Szilagyi H, He W, Goldman J, Zhao F. Engineering the Lymphatic Network: A Solution to Lymphedema. Adv Healthc Mater 2021; 10:e2001537. [PMID: 33502814 PMCID: PMC8483563 DOI: 10.1002/adhm.202001537] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/06/2020] [Indexed: 12/18/2022]
Abstract
Secondary lymphedema is a life-long disorder characterized by chronic tissue swelling and inflammation that obstruct interstitial fluid circulation and immune cell trafficking. Regenerating lymphatic vasculatures using various strategies represents a promising treatment for lymphedema. Growth factor injection and gene delivery have been developed to stimulate lymphangiogenesis and augment interstitial fluid resorption. Using bioengineered materials as growth factor delivery vehicles allows for a more precisely targeted lymphangiogenic activation within the injured site. The implantation of prevascularized lymphatic tissue also promotes in situ lymphatic capillary network formation. The engineering of larger scale lymphatic tissues, including lymphatic collecting vessels and lymph nodes constructed by bioengineered scaffolds or decellularized animal tissues, offers alternatives to reconnecting damaged lymphatic vessels and restoring lymph circulation. These approaches provide lymphatic vascular grafting materials to reimpose lymphatic continuity across the site of injury, without creating secondary injuries at donor sites. The present work reviews molecular mechanisms mediating lymphatic system development, approaches to promoting lymphatic network regeneration, and strategies for engineering lymphatic tissues, including lymphatic capillaries, collecting vessels, and nodes. Challenges of advanced translational applications are also discussed.
Collapse
Affiliation(s)
- Wenkai Jia
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77845
| | | | - Weilue He
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Jeremy Goldman
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77845
| |
Collapse
|
19
|
Li J, Xia Y, Fan X, Wu X, Yang F, Hu S, Wang Z. Extracellular vesicles derived from miR-199a-5p-modified adipose-derived mesenchymal stem cells alleviate immune thrombocytopenia by inhibiting T helper 17 differentiation. J Transl Med 2021; 101:318-327. [PMID: 33402685 DOI: 10.1038/s41374-020-00515-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/26/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
The abnormal differentiation of T helper 17 (Th17) cells is considered a vital promoter of immune thrombocytopenia (ITP) progression. Therefore, this study investigated the role of miR-199a-5p in Th17 differentiation and determined whether extracellular vesicles (EVs) derived from miR-199a-5p-modified adipose-derived mesenchymal stem cells (ADSCs) could relieve ITP by inhibiting Th17 differentiation. The miR-199a-5p level was lessened in the spleen tissues of mice with ITP, while the signal transducer and activator of transcription 3 (STAT3) expression and the population of Th17 in CD4+T cells were boosted. Functionally, miR-199a-5p overexpression lowered IL-17 secretion and the proportion of Th17/CD4+T cells. Further investigation showed that miR-199a-5p directly targeted STAT3 mRNA, and negatively modulated its expression. STAT3 overexpression was found to facilitate Th17 differentiation, which was subsequently abolished by miR-199a-5p overexpression. EVs isolated from miR-199a-5p-modified ADSCs (miR-199a-5p-EVs) highly expressed miR-199a-5p and could restrain CD4+T cells polarized toward a Th17 phenotype in vitro. Administering of miR-199a-5p-EVs elevated platelet counts and decreased the proportion of Th17/CD4+T cells in mice with ITP. Taken together, EVs derived from miR-199a-5p-modified ADSCs vividly repressed Th17 differentiation by transferring miR-199a-5p to CD4+T cells, thus ameliorating experimental ITP.
Collapse
Affiliation(s)
- Jianqin Li
- Hematology Department, The Children's Hospital of Soochow University, Suzhou, 215000, China.
| | - Yalin Xia
- Hematology Department, The Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Xiaoru Fan
- Hematology Department, The Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Xiaofang Wu
- Hematology Department, The Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Feiyun Yang
- Hematology Department, The Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Shaoyan Hu
- Hematology Department, The Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Zhaoyue Wang
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, 215000, China
| |
Collapse
|
20
|
Affiliation(s)
- Xianlei Li
- Department of Biomedical Engineering National University of Singapore 117583 Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering National University of Singapore 117583 Singapore
| | - Andy Tay
- Department of Biomedical Engineering National University of Singapore 117583 Singapore
- Institute for Health Innovation and Technology National University of Singapore 117599 Singapore
| |
Collapse
|
21
|
Marcozzi C, Frattini A, Borgese M, Rossi F, Barone L, Solari E, Valli R, Gornati R. Paracrine effect of human adipose-derived stem cells on lymphatic endothelial cells. Regen Med 2020; 15:2085-2098. [PMID: 33201769 DOI: 10.2217/rme-2020-0071] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The proposal of this study was to evaluate, in vitro, the potential paracrine effect of human adipose-derived stem cells (hASCs) to promote lymphangiogenesis in lymphatic endothelial cells isolated from rat diaphragmatic lymphatic vessels. Materials & methods: ELISA on VEGFA, VEGFC and IL6 in hASC-conditioned medium; LYVE1 immunostaining; and gene expression of PROX1, VEGFR3, VEGFC, VEGFA and IL6 were the methods used. Results: In 2D culture, hASC-conditioned medium was able to promote lymphatic endothelial cell survival, maintenance of endothelial cobblestone morphology and induction to form a vessel-like structure. Conclusion: The authors' results represent in vitro evidence of the paracrine effect of hASCs on lymphatic endothelial cells, suggesting the possible role of hASC-conditioned medium in developing new therapeutic approaches for lymphatic system-related dysfunction such as secondary lymphedema.
Collapse
Affiliation(s)
- Cristiana Marcozzi
- Department of Medicine & Surgery, Human Physiology, University of Insubria, 21100 Varese, Italy
| | - Annalisa Frattini
- Institute for Genetic & Biomedical Research, CNR, 20138 Milano, Italy.,Department of Medicine & Surgery, Human and Medical Genetics, University of Insubria, 21100 Varese, Italy
| | - Marina Borgese
- Department of Biotechnology & Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Federica Rossi
- Department of Biotechnology & Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Ludovica Barone
- Department of Biotechnology & Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Eleonora Solari
- Department of Medicine & Surgery, Human Physiology, University of Insubria, 21100 Varese, Italy
| | - Roberto Valli
- Department of Medicine & Surgery, Human and Medical Genetics, University of Insubria, 21100 Varese, Italy
| | - Rosalba Gornati
- Department of Biotechnology & Life Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
22
|
Li B, Yang J, Wang R, Li J, Li X, Zhou X, Qiu S, Weng R, Wu Z, Tang C, Li P. Delivery of vascular endothelial growth factor (VEGFC) via engineered exosomes improves lymphedema. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1498. [PMID: 33313243 PMCID: PMC7729376 DOI: 10.21037/atm-20-6605] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Lymphedema is a chronic disease results from impaired flow of the lymphatic system. Therefore, reconstruction of lymphatic system is crucial to treat limb lymphedema. Vascular endothelial growth factor (VEGFC) has been reported to be an important regulator involved in the growth and differentiation of lymphatic endothelial cells; however; the application of exosomes with VEGFC in the treatment of lymphedema has been rarely reported. Methods From the membrane-based fusion technology, we constructed engineered exosomes that overexpress CD63-VEGFC fusion protein (CD63-VEGFC/exos). We examined the in vitro effects of CD63-VEGFC/exos on the proliferation, migration, and tube formation of human dermal lymphatic endothelial cells (HDLECs) by MTT assay, migration assay, and tube formation assay, respectively. CD63-VEGFC/exos were embedded in sodium alginate hydrogel and their effect on lymphedema was evaluated by a mouse model. Results VEGFC could be successfully delivered to lymphatic endothelial cells via engineered CD63-VEGFC/exos. Treatment with CD63-VEGFC/exos resulted in a significant increase in the proliferation, migration, and tube formation of lymphatic endothelial cells. Using CD63-VEGFC/egos in sodium alginate hydrogel enabled a sequenced release of exosomes and markedly improved lymphedema in a mouse model. Conclusions Our findings supply a novel adipose tissue-derived stem cell (ADSC)-exo-based strategy that delivers target proteins to lymphatic endothelial cells and thus enhances the treatment of lymphedema.
Collapse
Affiliation(s)
- Bohan Li
- Department of Microsurgery & Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiantao Yang
- Department of Microsurgery & Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Raoping Wang
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jia Li
- Department of Microsurgery & Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xubo Li
- Department of Microsurgery & Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiang Zhou
- Department of Microsurgery & Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuai Qiu
- Department of Microsurgery & Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ricong Weng
- Department of Microsurgery & Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zichao Wu
- Department of Microsurgery & Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunyuan Tang
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ping Li
- Department of Microsurgery & Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Chen K, Sinelnikov MY, Reshetov IV, Timashev P, Gu Y, Mu L, Lu P, Zhang Y. Therapeutic Potential of Mesenchymal Stem Cells for Postmastectomy Lymphedema: A Literature Review. Clin Transl Sci 2020; 14:54-61. [PMID: 33460321 PMCID: PMC7877822 DOI: 10.1111/cts.12864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022] Open
Abstract
Upper limb lymphedema is one of the most common complications after breast cancer surgery and radiotherapy. Despite various physical therapy and surgical options available, the impaired lymph fluid drainage may be progressive due to lymphatic vascular insufficiency making treatment more difficulty. Stem cell therapy provides a promising alternative in the treatment of various chronic diseases. The wide applicability of cell therapy has been reviewed throughout literature. This review provides an overview of recent progress in the therapeutic effect of adult stem cells for primary and secondary lymphedema after breast surgery in preclinical studies and clinical cases. We start with a brief introduction about the pathophysiological mechanisms of postmastectomy lymphedema. Regarding existing treatments, we systematically summarize the benefits and limitations of recent progress. Because of their multidirectional differentiation potential and growth factor secretion, stem cell therapy shows promising results in the management of light to severe lymphedema. Increasing evidences have demonstrated a noticeable reduction in postmastectomy lymphedema and increased lymph-angiogenesis after specific stem cell therapy. Current data suggests that stem cell therapy in lymphedema treatment provides reversal of pathological reorganization associated with lymphedema progression. Finally, we propose potential strategies for overcoming the challenges in the development of multipotent progenitor cells for the treatment and prevention of lymphedema in clinical practice.
Collapse
Affiliation(s)
- Kuo Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Mikhail Y Sinelnikov
- Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Igor V Reshetov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Petr Timashev
- Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yuanting Gu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lan Mu
- Peking University People's Hospital, Beijing, China
| | - Pengwei Lu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| |
Collapse
|
24
|
Li ZJ, Yang E, Li YZ, Liang ZY, Huang JZ, Yu NZ, Long X. Application and prospect of adipose stem cell transplantation in treating lymphedema. World J Stem Cells 2020; 12:676-687. [PMID: 32843921 PMCID: PMC7415250 DOI: 10.4252/wjsc.v12.i7.676] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/08/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Lymphedema is a chronic, debilitating and incurable disease that affects 0.13%-2% of the global population. Emerging evidence indicates that adipose-derived stem cells (ADSCs) might serve as suitable seed cells for lymphatic tissue engineering and lymphedema therapy.
AIM To summarize applications of ADSCs for treating lymphedema in both animal studies and clinical trials.
METHODS A systematic search was performed on four databases – PubMed, Clinicaltrials.gov, the evidence-based Cochrane Library, and OVID – using the following search string: (“lymphedema” or “lymphoedema” or “lymphangiogenesis”) and (“adipose-derived stem cells” or “adipose-derived stromal cells” or “adipose-derived regenerative cells”). A manual search was performed by skimming the references of relevant studies. Animal studies and clinical trials using adipose-derived cells for the treatment of any kind of lymphedema were included.
RESULTS A total of eight research articles published before November 2019 were included for this analysis. Five articles focused on animal studies and another three focused on clinical trials. ADSC transplantation therapy was demonstrated to be effective against lymphedema in all studies. The animal studies found that coadministration of ADSCs and controlled-release vascular endothelial growth factor-C or platelet-rich plasma could improve the effectiveness of ADSC therapy. Three sequential clinical trials were conducted on breast cancer-related lymphedema patients, and all showed favorable results.
CONCLUSION ADSC-based therapy is a promising option for treating lymphedema. Large-scale, multicenter randomized controlled trials are needed to develop more effective and durable therapeutic strategies.
Collapse
Affiliation(s)
- Zhu-Jun Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Elan Yang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yun-Zhu Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zheng-Yun Liang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jiu-Zuo Huang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Nan-Ze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
25
|
Zhang S, Jin T, Wang L, Liu W, Zhang Y, Zheng Y, Lin Y, Yang M, He X, Lin H, Chen L, Tao J. Electro-Acupuncture Promotes the Differentiation of Endogenous Neural Stem Cells via Exosomal microRNA 146b After Ischemic Stroke. Front Cell Neurosci 2020; 14:223. [PMID: 32792909 PMCID: PMC7385414 DOI: 10.3389/fncel.2020.00223] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Background: Evidences indicate that exosomes-mediated delivery of microRNAs (miRNAs or miRs) is involved in the neurogenesis of stroke. This study was to investigate the role of exosomal miRNAs in non-drug therapy of electro-acupuncture (EA) regulating endogenous neural stem cells for stroke recovery. Methods: The model of focal cerebral ischemia and reperfusion in rats were established by middle cerebral artery occlusion (MCAO) and treated by EA. The exosomes were extracted from peri-ischemic striatum and identified by exosomal biomarkers, and detected differentially expressed miRNAs with microarray chip. Primary stem cells were cultured, and oxygen–glucose deprivation and reperfusion (OGD/R) was used to mimic vitro ischemic injury. Results: The levels of exosomal biomarkers TSG101 and CD81 were increased in peri-ischemic striatum after EA treatment, and we revealed 25 differentially expressed miRNAs in isolated exosomes, of which miR-146b was selected for further analysis, and demonstrated that EA increased miR-146b expression and its inhibitors could block the effects. Subsequently, we confirmed that EA upregulated miR-146b expression to promote neural stem cells differentiation into neurons in peri-ischemic striatum. In vitro, it was verified that OGD/R hindered neural stem cells differentiation, and miR-146b inhibitors furtherly suppressed its differentiation, simultaneously NeuroD1 was involved in neural stem cells differentiation into neurons. Moreover, in vivo we found EA promoted NeuroD1-mediated neural stem cells differentiation via miR-146b. In addition, EA also could improve neurological deficits through miR-146b after ischemic stroke. Conclusion: EA promotes the differentiation of endogenous neural stem cells via exosomal miR-146b to improve neurological injury after ischemic stroke.
Collapse
Affiliation(s)
- Shenghang Zhang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,The 900 Hospital of the Joint Logistic Team, Fuzhou, China
| | - Tingting Jin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lulu Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Weilin Liu
- Fujian University of Traditional Chinese Medicine, The Academy of Rehabilitation Industry, Fuzhou, China
| | - Yuhao Zhang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yi Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yunjiao Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Minguang Yang
- Fujian University of Traditional Chinese Medicine, The Academy of Rehabilitation Industry, Fuzhou, China
| | - Xiaojun He
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Huawei Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lidian Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
26
|
Bucan A, Dhumale P, Jørgensen MG, Dalaei F, Wiinholt A, Hansen CR, Hvidsten S, Baun C, Hejbøl EK, Schrøder HD, Sørensen JA. Comparison between stromal vascular fraction and adipose derived stem cells in a mouse lymphedema model. J Plast Surg Hand Surg 2020; 54:302-311. [PMID: 32520635 DOI: 10.1080/2000656x.2020.1772799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background: Lymphedema is one of the most common complications following breast cancer. Axillary lymph node dissection and radiotherapy are two well-known risk factors resulting in either removal or damage to the lymph nodes. As stem cells are known for their regenerative capabilities, they could theoretically repair/restore the damaged lymph vessels leading to a decrease in lymphedema.Methods: We evaluated the treatment of SVF and ASC on a mouse lymphedema model. Forty-five mice were allocated into three groups containing 15 mice each. The SVF group was injected with 100 μl containing 1 × 106 SVF, the ASC group with 100 μl ml containing 1 × 106 ASC and the NS with 100 μl ml of NS. Volumes of the mice were assessed weekly by μCT hindlimb volumetry for a total of 8 weeks. Lymph vessel morphometry was assessed by cross-sections of both hindlimbs stained for anti-LYVE1. Lymphatic function was assessed by lymphatic clearance.Results: The volume change between the groups was non-significant throughout all 8 weeks. The immunohistochemistry showed a statistically significant difference between the hindlimbs in ASC vs. NS group p = 0.032, 95% CI [-2121, -103].Conclusion: The volume of the hindlimbs showed that treatment with SVF or ASC yielded very similar results compared to the control group when assessed after 8 weeks. In week two the biggest difference between ASC and NS was seen but the difference diminished during the 8 weeks. The secondary outcomes showed that the lymph vessel lumen decreased when treated with ASC compared to the control group. Lymphoscintigraphy yielded non-significant results.
Collapse
Affiliation(s)
- Amar Bucan
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Pratibha Dhumale
- Department of Clinical Biochemistry and Pharmacology, University of Southern Denmark, Odense, Denmark
| | - Mads Gustaf Jørgensen
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Farima Dalaei
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Alexander Wiinholt
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Christian Rønn Hansen
- Laboratory of Radiation Physics, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Svend Hvidsten
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Christina Baun
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | | | | | - Jens Ahm Sørensen
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
27
|
Kamat P, Frueh FS, McLuckie M, Sanchez-Macedo N, Wolint P, Lindenblatt N, Plock JA, Calcagni M, Buschmann J. Adipose tissue and the vascularization of biomaterials: Stem cells, microvascular fragments and nanofat-a review. Cytotherapy 2020; 22:400-411. [PMID: 32507607 DOI: 10.1016/j.jcyt.2020.03.433] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/27/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
Tissue defects in the human body after trauma and injury require precise reconstruction to regain function. Hence, there is a great demand for clinically translatable approaches with materials that are both biocompatible and biodegradable. They should also be able to adequately integrate within the tissue through sufficient vascularization. Adipose tissue is abundant and easily accessible. It is a valuable tissue source in regenerative medicine and tissue engineering, especially with regard to its angiogenic potential. Derivatives of adipose tissue, such as microfat, nanofat, microvascular fragments, stromal vascular fraction and stem cells, are commonly used in research, but also clinically to enhance the vascularization of implants and grafts at defect sites. In plastic surgery, adipose tissue is harvested via liposuction and can be manipulated in three ways (macro-, micro- and nanofat) in the operating room, depending on its ultimate use. Whereas macro- and microfat are used as a filling material for soft tissue injuries, nanofat is an injectable viscous extract that primarily induces tissue remodeling because it is rich in growth factors and stem cells. In contrast to microfat that adds volume to a defect site, nanofat has the potential to be easily combined with scaffold materials due to its liquid and homogenous consistency and is particularly attractive for blood vessel formation. The same is true for microvascular fragments that are easily isolated from adipose tissue through collagenase digestion. In preclinical animal models, it has been convincingly shown that these vascular fragments inosculate with host vessels and subsequently accelerate scaffold perfusion and host tissue integration. Adipose tissue is also an ideal source of stem cells. It yields larger quantities of cells than any other source and is easier to access for both the patient and doctor compared with other sources such as bone marrow. They are often used for tissue regeneration in combination with biomaterials. Adipose-derived stem cells can be applied unmodified or as single cell suspensions. However, certain pretreatments, such as cultivation under hypoxic conditions or three-dimensional spheroids production, may provide substantial benefit with regard to subsequent vascularization in vivo due to induced growth factor production. In this narrative review, derivatives of adipose tissue and the vascularization of biomaterials are addressed in a comprehensive approach, including several sizes of derivatives, such as whole fat flaps for soft tissue engineering, nanofat or stem cells, their secretome and exosomes. Taken together, it can be concluded that adipose tissue and its fractions down to the molecular level promote, enhance and support vascularization of biomaterials. Therefore, there is a high potential of the individual fat component to be used in regenerative medicine.
Collapse
Affiliation(s)
- Pranitha Kamat
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland; Department of Plastic Surgery and Hand Surgery, University of Zurich, Zurich, Switzerland
| | - Florian S Frueh
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Michelle McLuckie
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Nadia Sanchez-Macedo
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Petra Wolint
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Nicole Lindenblatt
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Jan A Plock
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland; Department of Plastic Surgery and Hand Surgery, University of Zurich, Zurich, Switzerland
| | - Maurizio Calcagni
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Johanna Buschmann
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
28
|
Ahmadzadeh N, Robering JW, Kengelbach-Weigand A, Al-Abboodi M, Beier JP, Horch RE, Boos AM. Human adipose-derived stem cells support lymphangiogenesis in vitro by secretion of lymphangiogenic factors. Exp Cell Res 2020; 388:111816. [PMID: 31923426 DOI: 10.1016/j.yexcr.2020.111816] [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] [Received: 08/01/2019] [Revised: 12/22/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023]
Abstract
Lymphedema is a chronic progressive disease ultimately resulting in severe, disfiguring swelling and permanent changes of the affected tissues. Presently, there is no causal treatment approach of lymphedema. Therefore, most therapies are purely symptomatic. However, the recent use of stem cell-based therapies has offered new prospects for alternative treatment options. The present study was performed to investigate the effects of human adipose-derived stem cells (ADSCs) on human dermal lymphatic endothelial cells (HDLECs) in terms of basic in vitro lymphangiogenic assays (WST-8 assay, scratch assay, transmigration assay, sprouting assay, tube formation assay). The influence of ADSC-conditioned medium (ADSC-CM) on HDLECs was compared to recombinant VEGF-C, bFGF and HGF. Further ADSC-CM was characterized by protein microarray and enzyme-linked immunosorbent assay (ELISA). Although key-lymphangiogenic growth factors - like VEGF-C - could only be detected in low concentrations within the conditioned medium (CM), HDLECs were potently stimulated to proliferate, migrate and to form tube like structures by ADSC-CM. Despite concentrations more than hundredfold higher than those found in the conditioned medium, stimulation with recombinant VEGF-C, bFGF and HGF was still weaker compared to ADSC-CM. These results highlight the effectiveness of growth factors secreted by ADSC to stimulate HDLEC, potentially providing a promising new therapeutic approach for the treatment of lymphedema.
Collapse
Affiliation(s)
- Nima Ahmadzadeh
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Germany
| | - Jan W Robering
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Germany; Department of Plastic Surgery, Hand Surgery, Burns Center, University Hospital RWTH Aachen University, University Hospital Aachen, Germany
| | - Annika Kengelbach-Weigand
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Germany
| | - Majida Al-Abboodi
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Germany; Institute of Genetic Engineering and Biotechnology, University of Baghdad, Iraq
| | - Justus P Beier
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Germany; Department of Plastic Surgery, Hand Surgery, Burns Center, University Hospital RWTH Aachen University, University Hospital Aachen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Germany
| | - Anja M Boos
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Germany; Department of Plastic Surgery, Hand Surgery, Burns Center, University Hospital RWTH Aachen University, University Hospital Aachen, Germany.
| |
Collapse
|
29
|
Systemic Sclerosis Serum Significantly Impairs the Multi-Step Lymphangiogenic Process: in Vitro Evidence. Int J Mol Sci 2019; 20:ijms20246189. [PMID: 31817940 PMCID: PMC6940874 DOI: 10.3390/ijms20246189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/22/2019] [Accepted: 12/06/2019] [Indexed: 01/29/2023] Open
Abstract
In systemic sclerosis (SSc), the possible involvement of lymphatic microcirculation and lymphangiogenesis has traditionally been overshadowed by the greater emphasis placed on dysfunctional blood vascular system and angiogenesis. In the present in vitro study, we explore for the first time whether the SSc microenvironment may interfere with lymphangiogenesis, a complex, multi-step process in which lymphatic microvascular endothelial cells (LMVECs) sprout, migrate, and proliferate to generate new lymphatic capillaries. Normal human adult dermal LMVECs from three donors were treated with serum from SSc patients (n = 8), serum from healthy individuals (n = 8), or recombinant human vascular endothelial growth factor (VEGF)-C as a positive control for lymphangiogenesis. Cell proliferation, Boyden chamber Matrigel chemoinvasion, wound healing capacity, and lymphatic capillary morphogenesis on Geltrex were assayed. VEGF-C serum levels were measured by enzyme-linked immunosorbent assay. Gene and protein expression levels of the lymphangiogenic orchestrators VEGF receptor-3 (VEGFR-3)/Flt-4 and neuropilin-2 (NRP-2) were determined by real-time PCR and Western blotting, respectively. Conditioning with SSc serum significantly inhibited LMVEC proliferation, Matrigel invasion, and wound healing capacity with respect to healthy serum. The ability of LMVECs to form lymphatic tubes on Geltrex was also severely compromised in the presence of SSc serum. VEGF-C levels were comparable in SSc and healthy sera. Treatment with SSc serum resulted in a significant downregulation of both VEGFR-3/Flt-4 and NRP-2 mRNA and protein levels. In SSc, the pathologic environment severely hampers every lymphangiogenesis step, likely through the reduction of pro-lymphangiogenic VEGFR-3/NRP-2 co-receptor signaling. The impairment of the lymphangiogenic process opens a new scenario underlying SSc vascular pathophysiology, which is worth investigating further.
Collapse
|
30
|
Paracrine Effects of Adipose-Derived Stem Cells Promote Lymphangiogenesis in Irradiated Lymphatic Endothelial Cells. Plast Reconstr Surg 2019; 143:1189e-1200e. [PMID: 30907807 DOI: 10.1097/prs.0000000000005669] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND There is currently no reliable treatment for secondary lymphedema caused by lymph node dissection or radiotherapy; however, stem cell-based regenerative medicine is emerging as a promising remedy for such complications. The purpose of this study was to examine the effects of adipose-derived stem cells on lymphangiogenesis involving human dermal lymphatic endothelial cells exposed to ionizing radiation. METHODS Proliferation, migration, and tube formation were analyzed in human dermal lymphatic endothelial cells that were co-cultured with adipose-derived stem cells or cultured in adipose-derived stem cell-conditioned medium. The levels of lymphangiogenic factors secreted from adipose-derived stem cells were analyzed by enzyme-linked immunosorbent assays and Western blotting. RESULTS Co-culturing with adipose-derived stem cells and the use of adipose-derived stem cell-conditioned medium both significantly promoted proliferation, migration, and tube formation in nonirradiated human dermal lymphatic endothelial cells. The authors also found that irradiated adipose-derived stem cells had similar alleviative effects on irradiated human dermal lymphatic endothelial cells. Enzyme-linked immunosorbent assays and Western blotting analysis revealed that irradiating adipose-derived stem cells increased their secretion of basic fibroblast growth factor in a dose-dependent manner, whereas it caused no detectable change in their secretion of vascular endothelial growth factor A or C, or hepatocyte growth factor. CONCLUSIONS These results demonstrated that factors secreted by adipose-derived stem cells contribute to the promotion of lymphangiogenesis in irradiated human dermal lymphatic endothelial cells. The authors' findings also suggest that radiation potentiates the paracrine effects of adipose-derived stem cells by stimulating basic fibroblast growth factor protein expression.
Collapse
|
31
|
Chen CE, Chiang NJ, Perng CK, Ma H, Lin CH. Review of preclinical and clinical studies of using cell-based therapy for secondary lymphedema. J Surg Oncol 2019; 121:109-120. [PMID: 31385308 DOI: 10.1002/jso.25661] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/25/2019] [Indexed: 12/14/2022]
Abstract
Secondary lymphedema is associated with impaired lymph fluid drainage and remains incurable. Alternatively, cell-based therapy may pave the way for lymphedema treatment. We found 11 animal and seven human studies had been conducted from 2008 to 2018. Most studies showed great potential for this treatment modality. Emerging studies have focused on novel techniques, such as coupling cell therapy with lymph node transfer, or adding growth factors to cell therapy.
Collapse
Affiliation(s)
- Ching-En Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Nai-Jung Chiang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Department of Internal Medicine, Cheng Kung University Medical Center, Tainan, Taiwan
| | - Cherng-Kang Perng
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsu Ma
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Hsun Lin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
32
|
Tremp M, Sprenger L, Degrugillier L, Schaefer DJ, Madduri S, Schaeren S, Kalbermatten DF. Regeneration of nerve crush injury using adipose-derived stem cells: A multimodal comparison. Muscle Nerve 2019; 58:566-572. [PMID: 30028517 PMCID: PMC6646906 DOI: 10.1002/mus.26188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 01/26/2023]
Abstract
Introduction: To restore full function following nerve crush injuries is critical but challenging. In an attempt to develop a viable therapy, we evaluated the effect of rat adipose‐derived stem cells (rASC) in 2 different settings of a sciatic crush injury model. Methods: In the first group, after 14 days of nerve crush injury, rASCs were injected distal to the lesion under ultrasound guidance. In the other group, alleviation of compression through clip removal (CR) was combined with epineural injection of rASCs. Gait analyses, MRI, gastrocnemius muscle weight ratio (MWR), and histomorphometry were performed for outcome analysis. Results: CR combined with rASC injection resulted in less muscle atrophy, as evidenced by MWR. These findings are further supported by better functional and anatomical outcomes. Discussion: Animals treated with CR and epineural stem cell injection showed enhanced anatomical and functional recovery. Muscle Nerve58: 566–572, 2018
Collapse
Affiliation(s)
- Mathias Tremp
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Neuropathology, Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Lima Sprenger
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Neuropathology, Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Lucas Degrugillier
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Neuropathology, Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Dirk Johannes Schaefer
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Biomedical Engineering, Center for Bioengineering and Regenerative Medicine, University of Basel, Gewerbestrasse 14, 4123, Allschwil, Switzerland
| | - Srinivas Madduri
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Biomedical Engineering, Center for Bioengineering and Regenerative Medicine, University of Basel, Gewerbestrasse 14, 4123, Allschwil, Switzerland
| | - Stefan Schaeren
- Department of Spinal Surgery, University Hospital Basel, Basel, Switzerland
| | - Daniel Felix Kalbermatten
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital Basel, Basel, Switzerland.,Department of Neuropathology, Institute of Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
33
|
Sun Y, Lu B, Deng J, Jiang Z, Cao W, Dai T, Li S. IL-7 enhances the differentiation of adipose-derived stem cells toward lymphatic endothelial cells through AKT signaling. Cell Biol Int 2019; 43:394-401. [PMID: 30690788 DOI: 10.1002/cbin.11093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Our study was designed to investigate the effects of IL-7 during the differentiation process of adipose-derived stem cells (ADSCs) toward lymphatic endothelial cells (LECs). IL-7 was added to the traditional induced medium, which was called the IL-7 (+) group, while the group that used traditional induced medium was called the IL-7 (-) group. After 7 days of induction of ADSCs, a comprehensive analysis was conducted between these two groups. We examined the changes in Prox1, LYVE-1, Podoplanin and VEGFR-3 on the RNA and protein level and found that the expression of LEC markers in the IL-7 (+) group was higher than in the IL-7 (-) group. The characteristics of differentiated cells were confirmed by flow cytometry and immunofluorescence. At the same time, we detected the MAPK/ERK and PI3K/AKT pathway involved in the differentiation process, and we found that the phosphorylation of AKT increased, however the expression of ERK was not significantly changed. In conclusion, our study found that IL-7 could improve the differentiation efficiency of ADSCs toward LECs through AKT signaling pathways.
Collapse
Affiliation(s)
- Yiyu Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Bolun Lu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Jingcheng Deng
- Department of Plastic and Reconstructive Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Zhaohua Jiang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Weigang Cao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Tingting Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Shengli Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| |
Collapse
|
34
|
Kataru RP, Mehrara BJ, Kim H. Investigative strategies on lymphatic vessel modulation for treating lymphedema in future medicine. PRECISION AND FUTURE MEDICINE 2018. [DOI: 10.23838/pfm.2018.00142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
35
|
Meshram GG, Kaur N, Hura KS. Unilateral Primary Congenital Lymphedema of the Upper Limb in an 11-Month-Old Infant: A Clinical and Pharmacological Perspective. Open Access Maced J Med Sci 2018; 6:1682-1684. [PMID: 30337988 PMCID: PMC6182543 DOI: 10.3889/oamjms.2018.261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 08/21/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND: Lymphedema is the accumulation of a protein-rich fluid in the interstitial space due to reduced lymph transport capacity. Congenital primary lymphedema affecting only one of the upper limbs is a rarity. CASE REPORT: We present a case of an 11-month-old infant presenting with swelling of the right upper limb, which had gradually progressed over the past five months. Lymphoscintigraphy was suggestive of lymphatic blockade in the right upper limb. All other investigations were within normal limits. A diagnosis of primary congenital lymphedema affecting the right upper limb was made. The patient was managed conservatively with complex decongestive therapy and was requested regular follow-up. The lymphedema did not increase within four months of follow-up. CONCLUSIONS: Complex decongestive therapy is the cornerstone of the management of primary congenital lymphedema. New investigational therapies such as leukotriene B4 antagonists hold a promise for patients with lymphedema.
Collapse
Affiliation(s)
- Girish Gulab Meshram
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research and Dr Ram Manohar Lohia Hospital, New Delhi, India
| | - Neeraj Kaur
- Department of Radiology, The University of Texas Health Science Centre, San Antonio, Texas, USA
| | - Kanwaljeet Singh Hura
- Department of Paediatrics, Richmond University Medical Centre, Staten Island, New York, USA
| |
Collapse
|
36
|
Qian Y, Yang K, Mu L. [Research progress of vascularized lymph node transfer for extremity lymphedema]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:979-983. [PMID: 30238721 DOI: 10.7507/1002-1892.201801069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To review the current progress of clinical and experimental research of vascularized lymph node transfer for lymphedema. Methods The domestic and abroad literature about vascularized lymph node transfer in treatment of lymphedema was reviewed and analyzed. Results Experimental studies in animal model indicate that vascularized lymph node transfer can improve lymph node survival and show a promising effectiveness in reducing lymphedema. "Lymphatic wick" and "lymph pump" were the two main hypotheses proposed to explain the potential functional mechanism of vascularized lymph node transfer in treatment of lymphedema. Improvement in lymphedema symptoms are reported in most of the clinical trials, but the level of evidence to advocate this procedure in the treatment of lymphedema remains low because of the small number of the cases and problems in their methodologies. Conclusion Based on current evidence, vascularized lymph node transfer seems to be a promising treatment for lymphedema, but long-term well-designed studies are required to further explore the effectiveness of this procedure.
Collapse
Affiliation(s)
- Youlei Qian
- Department of Plastic & Medical Aesthetic Surgery, Peking University People's Hospital, Beijing, 100034, P.R.China
| | - Kai Yang
- Department of Plastic & Medical Aesthetic Surgery, Peking University People's Hospital, Beijing, 100034, P.R.China
| | - Lan Mu
- Department of Plastic & Medical Aesthetic Surgery, Peking University People's Hospital, Beijing, 100034,
| |
Collapse
|
37
|
Robering JW, Weigand A, Pfuhlmann R, Horch RE, Beier JP, Boos AM. Mesenchymal stem cells promote lymphangiogenic properties of lymphatic endothelial cells. J Cell Mol Med 2018; 22:3740-3750. [PMID: 29752774 PMCID: PMC6050462 DOI: 10.1111/jcmm.13590] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 12/30/2017] [Indexed: 12/18/2022] Open
Abstract
Lymphatic metastasis is one of the main prognostic factors concerning long-term survival of cancer patients. In this regard, the molecular mechanisms of lymphangiogenesis are still rarely explored. Also, the interactions between stem cells and lymphatic endothelial cells (LEC) in humans have not been well examined. Therefore, the main objective of this study was to assess the interactions between mesenchymal stem cells (MSC) and LEC using in vitro angiogenesis assays. Juvenile LEC were stimulated with VEGF-C, bFGF, MSC-conditioned medium (MSC-CM) or by co-culture with MSC. LEC proliferation was assessed using a MTT assay. Migration of the cells was determined with a wound healing assay and a transmigration assay. To measure the formation of lymphatic sprouts, LEC spheroids were embedded in collagen or fibrin gels. The LEC's capacity to form capillary-like structures was assessed by a tube formation assay on Matrigel® . The proliferation, migration and tube formation of LEC could be significantly enhanced by MSC-CM and by co-culture with MSC. The effect of stimulation with MSC-CM was stronger compared to stimulation with the growth factors VEGF-C and bFGF in proliferation and transmigration assays. Sprouting was stimulated by VEGF-C, bFGF and by MSC-CM. With this study, we demonstrate the potent stimulating effect of the MSC secretome on proliferation, migration and tube formation of LEC. This indicates an important role of MSC in lymphangiogenesis in pathological as well as physiological processes.
Collapse
Affiliation(s)
- Jan W Robering
- Department of Plastic and Hand Surgery, Laboratory of Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Annika Weigand
- Department of Plastic and Hand Surgery, Laboratory of Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Romy Pfuhlmann
- Department of Plastic and Hand Surgery, Laboratory of Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, Laboratory of Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Justus P Beier
- Department of Plastic and Hand Surgery, Laboratory of Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anja M Boos
- Department of Plastic and Hand Surgery, Laboratory of Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
38
|
Smolar J, Horst M, Sulser T, Eberli D. Bladder regeneration through stem cell therapy. Expert Opin Biol Ther 2018; 18:525-544. [DOI: 10.1080/14712598.2018.1439013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jakub Smolar
- Department of Urology, University Hospital Zurich, Schlieren, Switzerland
| | - Maya Horst
- Department of Urology, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Tulio Sulser
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Tendon Tissue Engineering: Mechanism and Effects of Human Tenocyte Coculture With Adipose-Derived Stem Cells. J Hand Surg Am 2018; 43:183.e1-183.e9. [PMID: 28888566 DOI: 10.1016/j.jhsa.2017.07.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/25/2017] [Accepted: 07/26/2017] [Indexed: 02/02/2023]
Abstract
PURPOSE Adipose-derived stem cells (ASCs) are a potential candidate for cell-based therapy targeting tendon injury; however, their therapeutic benefit relies on their ability to interact with native tenocytes. This study examines the mechanism and effects of coculturing human tenocytes and ASCs. METHODS Tenocytes (T) were directly cocultured with either ASCs (A) or fibroblasts (F) (negative control) in the following ratios: 50% T/50% A or F; 25% T/75% A or F; and 75% T/25% A or F. Cells were indirectly cocultured using a transwell insert that allowed for exchange of soluble factors only. Proliferation and collagen I production were measured and compared with monoculture controls. Synergy was quantified using the interaction index (II), which normalizes measured values by the expected values assuming no interaction (no synergy when II = 1). The ability of ASCs to elicit tenocyte migration was examined in vitro using a transwell migration assay and ex vivo using decellularized human flexor tendon explants. RESULTS Compared with monoculture controls, II of proliferation was greater than 1 for all tenocyte and ASC direct coculture ratios, but not for tenocyte and fibroblast direct coculture ratios or for tenocyte and ASC indirect coculture. The ASCs elicited greater tenocyte migration in vitro and ex vivo. The II of collagen I production was greater than 1 for direct coculture groups with 25% T/75% A and 75% T/25% A. CONCLUSIONS Direct coculture of ASCs and tenocytes demonstrated synergistic proliferation and collagen I production, and ASCs elicited tenocyte migration in vitro and ex vivo. These interactions play a key role in tendon healing and were absent when ASCs were replaced with fibroblasts, supporting the use of ASCs for cell-based therapy targeting tendon injuries. CLINICAL RELEVANCE When ASCs are delivered for cell-based therapy, they directly interact with native tenocytes to increase cell proliferation, collagen I production, and tenocyte migration, which may enhance tendon healing.
Collapse
|
40
|
Seo SK, Kim N, Lee JH, Kim SM, Lee SY, Bae JW, Hwang KK, Kim DW, Koch WJ, Cho MC. β-arrestin2 Affects Cardiac Progenitor Cell Survival through Cell Mobility and Tube Formation in Severe Hypoxia. Korean Circ J 2018; 48:296-309. [PMID: 29625512 PMCID: PMC5889979 DOI: 10.4070/kcj.2017.0119] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 01/03/2018] [Accepted: 01/17/2018] [Indexed: 11/15/2022] Open
Abstract
Background and Objectives β-arrestin2 (β-arr2) basically regulates multiple signaling pathways in mammalian cells by desensitization and internalization of G-protein coupled receptors (GPCRs). We investigated impacts of β-arr2 on survival, mobility, and tube formation of cardiac progenitor cells (CPCs) obtained from wild-type (WT) mouse (CPC-WT), and β-arr2 knock-out (KO) mouse (CPC-KO) cultured in presence or absence of serum and oxygen as non-canonical roles in GPCR system. Methods CPCs were cultured in Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 -based media containing fetal bovine serum and growth factors. Survival of 2 types of CPCs in hypoxia and/or serum deprivation was measured by fluorescence-activated cell sorting. Wound healing ability, and tube formation ability on Matrigel of 2 kinds of CPCs were compared in normoxic and hypoxic cultures. Protein expression related to survival and mobility were measured with the Western blot for each culture conditions. Results CPC-KO showed significantly worse mobility in the wound healing assay and in tube formation on Matrigel especially in hypoxic culture than did the CPC-WT. Also, CPC-KO showed significantly higher apoptosis fraction in both normoxic and hypoxic cultures than did the CPC-WT. Expression of proteins associated with cell survival and mobility, e.g., protein kinase B (Akt), β-catenin, and glycogen synthase kinase-3β (GSK-3β) was significantly worse in CPC-KO. Conclusions The CPC-KO had significantly worse cell mobility, tube formation ability, and survival than the CPC-WT, especially in the hypoxic cultures. Apparently, β-arr2 is important on CPC survival by means of mobility and tube formation in myocardial ischemia.
Collapse
Affiliation(s)
- Seul Ki Seo
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Nari Kim
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Ju Hee Lee
- Chungbuk Regional Cardiocerebrovascular Center, Chungbuk National University Hospital, Cheongju, Korea
| | - Sang Min Kim
- Chungbuk Regional Cardiocerebrovascular Center, Chungbuk National University Hospital, Cheongju, Korea
| | - Sang Yeub Lee
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea.,Chungbuk Regional Cardiocerebrovascular Center, Chungbuk National University Hospital, Cheongju, Korea
| | - Jang Whan Bae
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea. .,Chungbuk Regional Cardiocerebrovascular Center, Chungbuk National University Hospital, Cheongju, Korea
| | - Kyung Kuk Hwang
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea.,Chungbuk Regional Cardiocerebrovascular Center, Chungbuk National University Hospital, Cheongju, Korea
| | - Dong Woon Kim
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea.,Chungbuk Regional Cardiocerebrovascular Center, Chungbuk National University Hospital, Cheongju, Korea
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Myeong Chan Cho
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea.,Chungbuk Regional Cardiocerebrovascular Center, Chungbuk National University Hospital, Cheongju, Korea
| |
Collapse
|
41
|
Abstract
Solid tumor growth and metastasis require the interaction of tumor cells with the surrounding tissue, leading to a view of tumors as tissue-level phenomena rather than exclusively cell-intrinsic anomalies. Due to the ubiquitous nature of adipose tissue, many types of solid tumors grow in proximate or direct contact with adipocytes and adipose-associated stromal and vascular components, such as fibroblasts and other connective tissue cells, stem and progenitor cells, endothelial cells, innate and adaptive immune cells, and extracellular signaling and matrix components. Excess adiposity in obesity both increases risk of cancer development and negatively influences prognosis in several cancer types, in part due to interaction with adipose tissue cell populations. Herein, we review the cellular and noncellular constituents of the adipose "organ," and discuss the mechanisms by which these varied microenvironmental components contribute to tumor development, with special emphasis on obesity. Due to the prevalence of breast and prostate cancers in the United States, their close anatomical proximity to adipose tissue depots, and their complex epidemiologic associations with obesity, we particularly highlight research addressing the contribution of adipose tissue to the initiation and progression of these cancer types. Obesity dramatically modifies the adipose tissue microenvironment in numerous ways, including induction of fibrosis and angiogenesis, increased stem cell abundance, and expansion of proinflammatory immune cells. As many of these changes also resemble shifts observed within the tumor microenvironment, proximity to adipose tissue may present a hospitable environment to developing tumors, providing a critical link between adiposity and tumorigenesis. © 2018 American Physiological Society. Compr Physiol 8:237-282, 2018.
Collapse
Affiliation(s)
- Alyssa J. Cozzo
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ashley M. Fuller
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Liza Makowski
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
42
|
Wound Healing Effect of Conditioned Media Obtained From Adipose Tissue on Human Skin Cells: A Comparative in Vitro Study. Ann Plast Surg 2017; 77:156-63. [PMID: 25275476 DOI: 10.1097/sap.0000000000000358] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Split-thickness skin grafting is the gold standard to cover extensive acute and chronic wounds with a well-vascularized wound bed. Although some headway has been made in developing biological agents to speed up healing, there is still no treatment that sufficiently replaces skin grafts to date. The use of secretory factors of adipose tissue may be a feasible approach to developing topical wound applications for faster wound healing. METHODS In this study, the effect of conditioned media (CMs) of human adipose-derived stem cells (ASCs), adipocytes, or adipose tissue on human skin cells was evaluated for viability, proliferation, and migration in vitro. Differentiation potential of stem cells treated with CM was monitored by AdipoRed staining and qualitative real-time polymerase chain reaction. Angiogenic potential of human endothelial cells treated with CM was tested via sprouting assay. RESULTS The CM of adipose tissue significantly enhanced ASC proliferation (P < 0.01). Treatment with CM showed no inductive effect on ASC differentiation into adipocytes but, at the same time, significantly induced cell sprouting of endothelial cells (P < 0.001). We show for the first time that CM of adipose tissue is a potent inducer of proliferation of ASCs and angiogenesis, with comparable effects with those of stem cell-enriched CM. CONCLUSIONS We suggest the use of the secretome of adipose tissue to produce CM for topical application on wounds, rather than working with adipose tissue or including the difficult process of enriching the patients' stem cells in vitro.
Collapse
|
43
|
Intratendinous Injection of Hydrogel for Reseeding Decellularized Human Flexor Tendons. Plast Reconstr Surg 2017; 139:1305e-1314e. [PMID: 28538572 DOI: 10.1097/prs.0000000000003359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Decellularized cadaveric tendons are a potential source for reconstruction. Reseeding to enhance healing is ideal; however, cells placed on the tendon surface result in inadequate delivery. The authors used an injection technique to evaluate intratendinous cell delivery. METHODS Decellularized tendons were reseeded with adipose-derived stem cells in culture, and injected with fetal bovine serum or hydrogel. PKH26-stained cells in cross-section were quantified. To evaluate cell viability, the authors delivered luciferase-labeled cells and performed bioluminescent imaging. To evaluate synthetic ability, the authors performed immunohistochemistry of procollagen. Adipose-derived stem cells' ability to attract tenocytes was assessed using transwell inserts. Cell-to-cell interaction was assessed by co-culturing, measuring proliferation and collagen production, and quantifying synergy. Finally, tensile strength was tested. RESULTS Both fetal bovine serum (p < 0.001) and hydrogel (p < 0.001) injection led to more cells inside the tendon compared with culturing. Hydrogel injection initially demonstrated greater bioluminescence than culturing (p < 0.005) and fetal bovine serum injection (p < 0.05). Injection groups demonstrated intratendinous procollagen staining correlating with the cells' location. Co-culture led to greater tenocyte migration (p < 0.05). Interaction index of proliferation and collagen production assays were greater than 1 for all co-culture ratios, demonstrating synergistic proliferation and collagen production compared with controls (p < 0.05). There were no differences in tensile strength. CONCLUSIONS Hydrogel injection demonstrated the greatest intratendinous seeding efficiency and consistency, without compromising tensile strength. Intratendinous cells demonstrated synthetic capabilities and can potentially attract tenocytes inside the tendon, where synergy would promote intrinsic tendon healing. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
|
44
|
Adipose-Derived Stem Cells and Vascularized Lymph Node Transfers Successfully Treat Mouse Hindlimb Secondary Lymphedema by Early Reconnection of the Lymphatic System and Lymphangiogenesis. Plast Reconstr Surg 2017; 139:639-651. [PMID: 28234840 DOI: 10.1097/prs.0000000000003110] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Secondary lymphedema is often observed in postmalignancy treatment of the breast and the gynecologic organs, but effective therapies have not been established in chronic cases even with advanced physiologic operations. Currently, reconstructive surgery with novel approaches has been attempted. METHODS The hindlimbs of 10-week-old male C57BL/6J mice, after 30-Gy x-irradiation, surgical lymph node dissection, and 5-mm gap creation, were divided into four groups, with vascularized lymph node transfer abdominal flap and 1.0 × 10 adipose-derived stem cells. Lymphatic flow assessment, a water-displacement plethysmometer paw volumetry test, tissue quantification of lymphatic vessels, and functional analysis of lymphatic vessels and nodes were performed. RESULTS Photodynamic Eye images, using indocyanine green fluorescence, demonstrated immediate staining in subiliac lymph nodes, and linear pattern imaging of the proximal region was observed with the combined treatment of adipose-derived stem cells and vascularized lymph node transfer. Both percentage improvement and percentage deterioration with the combined treatment of adipose-derived stem cells and vascularized lymph node transfer were significantly better than with other treatments (p < 0.05). The numbers of lymphatic vessels with LYVE-1 immunoreactivity significantly increased in mice treated with adipose-derived stem cells (p < 0.05), and B16 melanoma cells were metastasized in groups treated with vascularized lymph node transfers by day 28. CONCLUSIONS Adipose-derived stem cells increase the number of lymphatic vessels and vascularized lymph node transfers induce the lymphatic flow drainage to the circulatory system. Combined adipose-derived stem cell and vascularized lymph node transfer treatment in secondary lymphedema may effectively decrease edema volume and restore lymphatic function by lymphangiogenesis and the lymphatic-to-venous circulation route.
Collapse
|
45
|
Knezevic L, Schaupper M, Mühleder S, Schimek K, Hasenberg T, Marx U, Priglinger E, Redl H, Holnthoner W. Engineering Blood and Lymphatic Microvascular Networks in Fibrin Matrices. Front Bioeng Biotechnol 2017; 5:25. [PMID: 28459049 PMCID: PMC5394507 DOI: 10.3389/fbioe.2017.00025] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/28/2017] [Indexed: 01/20/2023] Open
Abstract
Vascular network engineering is essential for nutrient delivery to tissue-engineered constructs and, consequently, their survival. In addition, the functionality of tissues also depends on tissue drainage and immune cell accessibility, which are the main functions of the lymphatic system. Engineering both the blood and lymphatic microvasculature would advance the survival and functionality of tissue-engineered constructs. The aim of this study was to isolate pure populations of lymphatic endothelial cells (LEC) and blood vascular endothelial cells (BEC) from human dermal microvascular endothelial cells and to study their network formation in our previously described coculture model with adipose-derived stromal cells (ASC) in fibrin scaffolds. We could follow the network development over a period of 4 weeks by fluorescently labeling the cells. We show that LEC and BEC form separate networks, which are morphologically distinguishable and sustainable over several weeks. In addition, lymphatic network development was dependent on vascular endothelial growth factor (VEGF)-C, resulting in denser networks with increasing VEGF-C concentration. Finally, we confirm the necessity of cell–cell contact between endothelial cells and ASC for the formation of both blood and lymphatic microvascular networks. This model represents a valuable platform for in vitro drug testing and for the future in vivo studies on lymphatic and blood microvascularization.
Collapse
Affiliation(s)
- Lea Knezevic
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mira Schaupper
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Severin Mühleder
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Katharina Schimek
- Technische Universität Berlin, Medical Biotechnology, Berlin, Germany.,TissUse GmbH, Berlin, Germany
| | | | | | - Eleni Priglinger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Wolfgang Holnthoner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
46
|
Ran S, Wilber A. Novel role of immature myeloid cells in formation of new lymphatic vessels associated with inflammation and tumors. J Leukoc Biol 2017; 102:253-263. [PMID: 28408396 DOI: 10.1189/jlb.1mr1016-434rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 03/14/2017] [Accepted: 03/17/2017] [Indexed: 12/18/2022] Open
Abstract
Inflammation triggers an immune cell-driven program committed to restoring homeostasis to injured tissue. Central to this process is vasculature restoration, which includes both blood and lymphatic networks. Generation of new vessels or remodeling of existing vessels are also important steps in metastasis-the major cause of death for cancer patients. Although roles of the lymphatic system in regulation of inflammation and cancer metastasis are firmly established, the mechanisms underlying the formation of new lymphatic vessels remain a subject of debate. Until recently, generation of new lymphatics in adults was thought to occur exclusively through sprouting of existing vessels without help from recruited progenitors. However, emerging findings from clinical and experimental studies show that lymphoendothelial progenitors, particularly those derived from immature myeloid cells, play an important role in this process. This review summarizes current evidence for the existence and significant roles of myeloid-derived lymphatic endothelial cell progenitors (M-LECPs) in generation of new lymphatics. We describe specific markers of M-LECPs and discuss their biologic behavior in culture and in vivo, as well as currently known molecular mechanisms of myeloid-lymphatic transition (MLT). We also discuss the implications of M-LECPs for promoting adaptive immunity, as well as cancer metastasis. We conclude that improved mechanistic understanding of M-LECP differentiation and its role in adult lymphangiogenesis may lead to new therapeutic approaches for correcting lymphatic insufficiency or excessive formation of lymphatic vessels in human disorders.
Collapse
Affiliation(s)
- Sophia Ran
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, and Simmons Cancer Institute, Springfield, Illinois, USA
| | - Andrew Wilber
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, and Simmons Cancer Institute, Springfield, Illinois, USA
| |
Collapse
|
47
|
Smolar J, Salemi S, Horst M, Sulser T, Eberli D. Stem Cells in Functional Bladder Engineering. Transfus Med Hemother 2016; 43:328-335. [PMID: 27781020 PMCID: PMC5073506 DOI: 10.1159/000447977] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/23/2016] [Indexed: 12/24/2022] Open
Abstract
Conditions impairing bladder function in children and adults, such as myelomeningocele, posterior urethral valves, bladder exstrophy or spinal cord injury, often need urinary diversion or augmentation cystoplasty as when untreated they may cause severe bladder dysfunction and kidney failure. Currently, the gold standard therapy of end-stage bladder disease refractory to conservative management is enterocystoplasty, a surgical enlargement of the bladder with intestinal tissue. Despite providing functional improvement, enterocystoplasty is associated with significant long-term complications, such as recurrent urinary tract infections, metabolic abnormalities, stone formation, and malignancies. Therefore, there is a strong clinical need for alternative therapies for these reconstructive procedures, of which stem cell-based tissue engineering (TE) is considered to be the most promising future strategy. This review is focused on the recent progress in bladder stem cell research and therapy and the challenges that remain for the development of a functional bladder wall.
Collapse
Affiliation(s)
- Jakub Smolar
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Souzan Salemi
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Maya Horst
- Division of Pediatric Urology, Department of Pediatric Surgery, University Children's Hospital, Zurich, Switzerland
| | - Tullio Sulser
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Daniel Eberli
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
48
|
Adipose-Derived Stem Cells Promote Peripheral Nerve Regeneration In Vivo without Differentiation into Schwann-Like Lineage. Plast Reconstr Surg 2016; 137:318e-330e. [PMID: 26818322 DOI: 10.1097/01.prs.0000475762.86580.36] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND During recent decades, multipotent stem cells were found to reside in the adipose tissue, and these adipose-derived stem cells were shown to play beneficial roles, like those of Schwann cells, in peripheral nerve regeneration. However, it has not been well established whether adipose-derived stem cells offer beneficial effects to peripheral nerve injuries in vivo as Schwann cells do. Furthermore, the in situ survival and differentiation of adipose-derived stem cells after transplantation at the injured peripheral nerve tissue remain to be fully elucidated. METHODS Adipose-derived stem cells and Schwann cells were transplanted with gelatin hydrogel tubes at the artificially blunted sciatic nerve lesion in mice. Neuroregenerative abilities of them were comparably estimated. Cre-loxP-mediated fate tracking was performed to visualize survival in vivo of transplanted adipose-derived stem cells and to investigate whether they differentiated into Schwann linage cells at the peripheral nerve injury site. RESULTS The transplantation of adipose-derived stem cells promoted regeneration of axons, formation of myelin, and restoration of denervation muscle atrophy to levels comparable to those achieved by Schwann cell transplantation. The adipose-derived stem cells survived for at least 4 weeks after transplantation without differentiating into Schwann cells. CONCLUSIONS Transplanted adipose-derived stem cells did not differentiate into Schwann cells but promoted peripheral nerve regeneration at the injured site. The neuroregenerative ability was comparable to that of Schwann cells. Adipose-derived stem cells at an undifferentiated stage may be used as an alternative cell source for autologous cell therapy for patients with peripheral nerve injury.
Collapse
|
49
|
Yoshida S, Hamuy R, Hamada Y, Yoshimoto H, Hirano A, Akita S. Adipose-derived stem cell transplantation for therapeutic lymphangiogenesis in a mouse secondary lymphedema model. Regen Med 2016; 10:549-62. [PMID: 26237700 DOI: 10.2217/rme.15.24] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM Secondary lymphedema is observed in common after postmalignancy treatment of the breast and the gynecologic organs but effective therapies are not established. Adipose-derived stem cells (ADSCs), which are pluripotent, regenerative in local injection, are tested for murine hindlimb secondary lymphedema by regenerative method. METHODS & RESULTS Mice were divided into four groups: no ADSCs, 1 × 10(6) ADSCs, 1 × 10(5) ADSCs and 1 × 10(4) ADSCs (each group, n = 20) in a stringent surgical resection and irradiation. Circumferential measurement, lymphatic flow assessment and quantification of lymphatic vessels were performed. RESULTS The numbers of lymphatic vessels by LYVE-1 immunohistochemistry, and VEGF-C- or VEGFR3-expressing cells were significantly increased in transplanted groups (p < 0.05). CONCLUSION ADSCs can restore the lymphatic vascular network in secondary lymphedema with increased collecting vessels.
Collapse
Affiliation(s)
- Shuhei Yoshida
- Division of Plastic & Reconstructive Surgery, Department of Developmental & Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852 8501, Japan
| | - Rodrigo Hamuy
- Division of Plastic & Reconstructive Surgery, Department of Developmental & Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852 8501, Japan
| | - Yuuichi Hamada
- Division of Plastic & Reconstructive Surgery, Oita Sanai Medical Center, 1213 Ichi, Oita 870-1151, Japan
| | - Hiroshi Yoshimoto
- Division of Plastic & Reconstructive Surgery, Department of Developmental & Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852 8501, Japan
| | - Akiyoshi Hirano
- Division of Plastic & Reconstructive Surgery, Department of Developmental & Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852 8501, Japan
| | - Sadanori Akita
- Division of Plastic & Reconstructive Surgery, Department of Developmental & Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852 8501, Japan
| |
Collapse
|
50
|
Strassburg S, Torio-Padron N, Finkenzeller G, Frankenschmidt A, Stark G. Adipose-Derived Stem Cells Support Lymphangiogenic Parameters In Vitro. J Cell Biochem 2016; 117:2620-9. [DOI: 10.1002/jcb.25557] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 03/24/2016] [Indexed: 12/12/2022]
Affiliation(s)
- S. Strassburg
- Department of Plastic and Hand Surgery; University of Freiburg Medical Center; Hugstetter Str. 55 Freiburg 79106 Germany
| | - N. Torio-Padron
- Department of Plastic and Hand Surgery; University of Freiburg Medical Center; Hugstetter Str. 55 Freiburg 79106 Germany
| | - G. Finkenzeller
- Department of Plastic and Hand Surgery; University of Freiburg Medical Center; Hugstetter Str. 55 Freiburg 79106 Germany
| | - A. Frankenschmidt
- Department of Urologie; University of Freiburg Medical Center; Hugstetter Str. 55 Freiburg 79106 Germany
| | - G.B. Stark
- Department of Plastic and Hand Surgery; University of Freiburg Medical Center; Hugstetter Str. 55 Freiburg 79106 Germany
| |
Collapse
|