1
|
Kong AM, Idris ZA, Urrutia-Cabrera D, Lees JG, Phang RJ, Mitchell GM, Wong RC, Lim SY. NOS3 regulates angiogenic potential of human induced pluripotent stem cell-derived endothelial cells. Biochem Biophys Rep 2024; 40:101876. [PMID: 39634339 PMCID: PMC11616527 DOI: 10.1016/j.bbrep.2024.101876] [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: 01/23/2024] [Revised: 08/12/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
Incorporation of blood capillaries in engineered tissues and their functional connection to host blood vessels is essential for success in engineering vascularized tissues, a process which involves spatial patterning of endothelial cells (ECs) to form functional and integrated vascular networks. Different types of ECs have been employed for vascular network formation and each source offers advantages and disadvantages. While ECs derived from induced pluripotent stem cells (iPSC-ECs) offer advantages over primary ECs in that they can be generated in large quantities for autologous applications, their suitability for disease modelling and cell replacement therapies is not well-explored. The present study compares the angiogenic capacity of iPSC-ECs and primary ECs (cardiac microvascular ECs and lymphatic microvascular ECs) using an in vitro tubulogenesis assay, revealing comparable performance in forming a pseudo-capillary network on Matrigel. Analysis of genes encoding angiogenic factors (VEGFA, VEGFC, VEGFD and ANG), endothelial cell markers (PECAM1, PCDH12 and NOS3) and proliferation markers (AURKB and MKI67) indicates a significant positive correlation between NOS3 mRNA expression levels and various tubulogenic parameters. Further experimentation using a CRISPR activation system demonstrates a positive impact of NOS3 on tubulogenic activity of ECs, suggesting that iPSC-ECs can be enhanced with endogenous NOS3 activation. Collectively, these findings highlight the potential of iPSC-ECs in generating vascularized tissues and advancing therapeutic vascularization.
Collapse
Affiliation(s)
- Anne M. Kong
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
| | - Zulhusni A. Idris
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
- School of Engineering, University of Melbourne, VIC, Australia
| | - Daniel Urrutia-Cabrera
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Jarmon G. Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
| | - Ren Jie Phang
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
| | - Geraldine M. Mitchell
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
- Faculty of Health Sciences, Australian Catholic University, VIC, Australia
| | - Raymond C.B. Wong
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Shiang Y. Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, VIC, Australia
- Departments of Surgery, Ophthalmology and Medicine, University of Melbourne, VIC, Australia
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Victoria, Monash University, Australia
- National Heart Research Institute Singapore, National Heart Centre, Singapore
| |
Collapse
|
2
|
Vu BT, Le HT, Nguyen KN, Van Pham P. Hypoxia, Serum Starvation, and TNF-α Can Modify the Immunomodulation Potency of Human Adipose-Derived Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021:3-18. [PMID: 34739720 DOI: 10.1007/5584_2021_672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Adipose-derived stem cells (ADSCs) are mesenchymal stem cells (MSCs) that are found in adipose tissues, which are easily obtained from liposuction procedures using an enzyme mixture. The adhering cells are then selectively cultivated. ADSCs have great potential in regenerative medicine because they are plentiful, easily accessible, and less invasive. They also have an impressive proliferation ability and can be differentiated into mesenchymal lineages and trans-differentiating into many other cell types. In particular, they have extraordinary abilities in immunomodulation. This study aimed to investigate the effects of culture conditions (hypoxia, starvation, and TNF-α treatment) on the immunomodulation of human ADSCs. METHODS Human ADSCs were expanded in vitro in the standard condition before they were cultured in different stress conditions. ADSCs from passages fifth were confirmed as MSCs by some standard assays suggested by the International Society for Cell and Gene Therapy. These MSCs were used to culture in four different stress conditions: hypoxia, serum starvation, and TNF-α treatment in 48 h. After treatments, MSCs were used to evaluate their immunomodulation capacity using MSCs mixed lymphocyte reaction assay, and the concentrations of IDO, PGE2, IL-6, and IL-10 were secreted in the culture medium. RESULTS In different stress conditions, ADSCs exhibited different responses related to their immunomodulation. In serum starvation, ADSCs exerted a strong secretion of IDO and PGE2, whereas they showed strong IL-6 secretion in the TNF-α-supplemented medium. When exposed to lymphocytes, ADSCs caused an increase in the ratio of regulatory T cells (Tregs), and co-culture lymphocytes with ADSCs induced in hypoxic malnutrition conditions increased the IL-10 level the most. In addition, when exposed to dendritic cells (DCs), ADSCs inhibited the mature marker expressions of the DCs. CONCLUSION The current research showed that ADSCs change their immunomodulation properties to survive in in vitro culture environments. Treatment of ADSCs in the starvation medium for 48 h can increase the immunomodulation of ADSCs.
Collapse
Affiliation(s)
- Binh Thanh Vu
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Hanh Thi Le
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Stem Cell Institute, University of Science, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Khanh Nha Nguyen
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Stem Cell Institute, University of Science, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Phuc Van Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam.
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam.
- Stem Cell Institute, University of Science, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam.
| |
Collapse
|
3
|
Jeannerat A, Peneveyre C, Armand F, Chiappe D, Hamelin R, Scaletta C, Hirt-Burri N, de Buys Roessingh A, Raffoul W, Applegate LA, Laurent A. Hypoxic Incubation Conditions for Optimized Manufacture of Tenocyte-Based Active Pharmaceutical Ingredients of Homologous Standardized Transplant Products in Tendon Regenerative Medicine. Cells 2021; 10:cells10112872. [PMID: 34831095 PMCID: PMC8616528 DOI: 10.3390/cells10112872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022] Open
Abstract
Human fetal progenitor tenocytes (hFPT) produced in defined cell bank systems have recently been characterized and qualified as potential therapeutic cell sources in tendon regenerative medicine. In view of further developing the manufacture processes of such cell-based active pharmaceutical ingredients (API), the effects of hypoxic in vitro culture expansion on key cellular characteristics or process parameters were evaluated. To this end, multiple aspects were comparatively assessed in normoxic incubation (i.e., 5% CO2 and 21% O2, standard conditions) or in hypoxic incubation (i.e., 5% CO2 and 2% O2, optimized conditions). Experimentally investigated parameters and endpoints included cellular proliferation, cellular morphology and size distribution, cell surface marker panels, cell susceptibility toward adipogenic and osteogenic induction, while relative protein expression levels were analyzed by quantitative mass spectrometry. The results outlined conserved critical cellular characteristics (i.e., cell surface marker panels, cellular phenotype under chemical induction) and modified key cellular parameters (i.e., cell size distribution, endpoint cell yields, matrix protein contents) potentially procuring tangible benefits for next-generation cell manufacturing workflows. Specific proteomic analyses further shed some light on the cellular effects of hypoxia, potentially orienting further hFPT processing for cell-based, cell-free API manufacture. Overall, this study indicated that hypoxic incubation impacts specific hFPT key properties while preserving critical quality attributes (i.e., as compared to normoxic incubation), enabling efficient manufacture of tenocyte-based APIs for homologous standardized transplant products.
Collapse
Affiliation(s)
- Annick Jeannerat
- Applied Research Department, LAM Biotechnologies SA, CH-1066 Épalinges, Switzerland; (A.J.); (C.P.)
| | - Cédric Peneveyre
- Applied Research Department, LAM Biotechnologies SA, CH-1066 Épalinges, Switzerland; (A.J.); (C.P.)
| | - Florence Armand
- Proteomics Core Facility and Technology Platform, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland; (F.A.); (D.C.); (R.H.)
| | - Diego Chiappe
- Proteomics Core Facility and Technology Platform, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland; (F.A.); (D.C.); (R.H.)
| | - Romain Hamelin
- Proteomics Core Facility and Technology Platform, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland; (F.A.); (D.C.); (R.H.)
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (C.S.); (N.H.-B.); (L.A.A.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (C.S.); (N.H.-B.); (L.A.A.)
| | - Anthony de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Wassim Raffoul
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (C.S.); (N.H.-B.); (L.A.A.)
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Alexis Laurent
- Applied Research Department, LAM Biotechnologies SA, CH-1066 Épalinges, Switzerland; (A.J.); (C.P.)
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (C.S.); (N.H.-B.); (L.A.A.)
- Manufacturing Department, TEC-PHARMA SA, CH-1038 Bercher, Switzerland
- Correspondence: ; Tel.: +41-21-546-42-00
| |
Collapse
|
4
|
The Role of Oxidative Stress in Cardiac Disease: From Physiological Response to Injury Factor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5732956. [PMID: 32509147 PMCID: PMC7244977 DOI: 10.1155/2020/5732956] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/11/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are highly reactive chemical species containing oxygen, controlled by both enzymatic and nonenzymatic antioxidant defense systems. In the heart, ROS play an important role in cell homeostasis, by modulating cell proliferation, differentiation, and excitation-contraction coupling. Oxidative stress occurs when ROS production exceeds the buffering capacity of the antioxidant defense systems, leading to cellular and molecular abnormalities, ultimately resulting in cardiac dysfunction. In this review, we will discuss the physiological sources of ROS in the heart, the mechanisms of oxidative stress-related myocardial injury, and the implications of experimental studies and clinical trials with antioxidant therapies in cardiovascular diseases.
Collapse
|
5
|
Beigi Boroujeni F, Pasbakhsh P, Mortezaee K, Pirhajati V, Alizadeh R, Aryanpour R, Madadi S, Ragerdi Kashani I. Intranasal delivery of SDF-1α-preconditioned bone marrow mesenchymal cells improves remyelination in the cuprizone-induced mouse model of multiple sclerosis. Cell Biol Int 2019; 44:499-511. [PMID: 31631484 DOI: 10.1002/cbin.11250] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS) that leads to disability in middle-aged individuals. High rates of apoptosis and inappropriate homing are limitations for the application of stem cells in cell therapy. Preconditioning of bone marrow mesenchymal stem cells (BMSCs) with stromal cell-derived factor 1α (SDF-1α), also called C-X-C motif chemokine 12 (CXCL12), is an approach for improving the functional features of the cells. The aim of this study was to investigate the therapeutic efficacy of intranasal delivery of SDF-1α preconditioned BMSCs in the cuprizone-induced chronically demyelinated mice model. BMSCs were isolated, cultured, and preconditioned with SDF-1α. Then, intranasal delivery of the preconditioned cells was performed in the C57BL/6 mice receiving cuprizone for 12 weeks. Animals were killed at 30 days after cell delivery. SDF-1α preconditioning increased C-X-C chemokine receptor type 4 (CXCR4) expression on the surface of BMSCs, improved survival of the cells, and decreased their apoptosis in vitro. SDF-1α preconditioning also improved CXCL12 level within the brain, and enhanced spatial learning and memory (assessed by Morris water maze [MWM]), and myelination (assessed by Luxol fast blue [LFB] and transmission electron microscopy [TEM]). In addition, preconditioning of BMSCs with SDF-1α reduced the protein expressions of glial fibrillary acidic protein and ionized calcium-binding adapter molecule (Iba-1) and increased the expressions of oligodendrocyte lineage transcription factor-2 (Olig-2) and adenomatous polyposis coli (APC), evaluated by immunofluorescence. The results showed the efficacy of intranasal delivery of SDF-1α-preconditioned BMSCs for improving remyelination in the cuprizone model of MS.
Collapse
Affiliation(s)
- Fatemeh Beigi Boroujeni
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Science, Tehran, 1417653761, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Science, Tehran, 1417653761, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Vahid Pirhajati
- Neuroscience Research Center, Vice-Chancellor for Research and Technology, Iran University of Medical Science, Tehran, 1449614535, Iran
| | - Rafieh Alizadeh
- ENT and Head & Neck Research Center and Department, Hazrat Rasoul Akram Hospital, Tehran, 1445613131, Iran
| | - Roya Aryanpour
- Department of Anatomy, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, 7591741417, Iran
| | - Soheila Madadi
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Science, Tehran, 1417653761, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Science, Tehran, 1417653761, Iran
| |
Collapse
|
6
|
Hummitzsch L, Zitta K, Rusch R, Cremer J, Steinfath M, Gross J, Fandrich F, Berndt R, Albrecht M. Characterization of the Angiogenic Potential of Human Regulatory Macrophages (Mreg) after Ischemia/Reperfusion Injury In Vitro. Stem Cells Int 2019; 2019:3725863. [PMID: 31341483 PMCID: PMC6614961 DOI: 10.1155/2019/3725863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022] Open
Abstract
Ischemia/reperfusion- (I/R-) induced organ damage represents one of the main causes of death worldwide, and new strategies to reduce I/R injury are urgently needed. We have shown that programmable cells of monocytic origin (PCMO) respond to I/R with the release of angiogenic mediators and that transplantation of PCMO results in increased neovascularization. Human regulatory macrophages (Mreg), which are also of monocytic origin, have been successfully employed in clinical transplantation studies due to their immunomodulatory properties. Here, we investigated whether Mreg also possess angiogenic potential in vitro and could represent a treatment option for I/R-associated illnesses. Mreg were differentiated using peripheral blood monocytes from different donors (N = 14) by incubation with M-CSF and human AB serum and stimulation with INF-gamma. Mreg cultures were subjected to 3 h of hypoxia and 24 h of reoxygenation (resembling I/R) or the respective nonischemic control. Cellular resilience, expression of pluripotency markers, secretion of angiogenic proteins, and influence on endothelial tube formation as a surrogate marker for angiogenesis were investigated. Mreg showed resilience against I/R that did not lead to increased cell damage. Mreg express DHRS9 as well as IDO and display a moderate to low expression pattern of several pluripotency genes (e.g., NANOG, OCT-4, and SOX2). I/R resulted in an upregulation of IDO (p < 0.001) while C-MYC and KLF4 were downregulated (p < 0.001 and p < 0.05). Proteome profiling revealed the secretion of numerous angiogenic proteins by Mreg of which several were strongly upregulated by I/R (e.g., MIP-1alpha, 19.9-fold; GM-CSF, 19.2-fold; PTX3, 5.8-fold; IL-1β, 5.2-fold; and MCP-1, 4.7-fold). The angiogenic potential of supernatants from Mreg subjected to I/R remains inconclusive. While Mreg supernatants from 3 donors induced tube formation, 2 supernatants were not effective. We suggest that Mreg may prove beneficial as a cell therapy-based treatment option for I/R-associated illnesses. However, donor characteristics seem to crucially influence the effectiveness of Mreg treatment.
Collapse
Affiliation(s)
- Lars Hummitzsch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Karina Zitta
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Rene Rusch
- Department of Cardiovascular Surgery, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Jochen Cremer
- Department of Cardiovascular Surgery, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Markus Steinfath
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Justus Gross
- Clinic for Vascular Surgery, Bad Segeberg, Germany
| | - Fred Fandrich
- Department of Applied Cell Therapy, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Rouven Berndt
- Department of Cardiovascular Surgery, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Martin Albrecht
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
7
|
Cacciotti I, Ciocci M, Di Giovanni E, Nanni F, Melino S. Hydrogen Sulfide-Releasing Fibrous Membranes: Potential Patches for Stimulating Human Stem Cells Proliferation and Viability under Oxidative Stress. Int J Mol Sci 2018; 19:E2368. [PMID: 30103516 PMCID: PMC6121677 DOI: 10.3390/ijms19082368] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/07/2018] [Accepted: 08/08/2018] [Indexed: 01/04/2023] Open
Abstract
The design of biomaterial platforms able to release bioactive molecules is mandatory in tissue repair and regenerative medicine. In this context, electrospinning is a user-friendly, versatile and low-cost technique, able to process different kinds of materials in micro- and nano-fibers with a large surface area-to-volume ratio for an optimal release of gaseous signaling molecules. Recently, the antioxidant and anti-inflammatory properties of the endogenous gasotramsmitter hydrogen sulfide (H₂S), as well as its ability to stimulate relevant biochemical processes on the growth of mesenchymal stem cells (MSC), have been investigated. Therefore, in this work, new poly(lactic) acid fibrous membranes (PFM), doped and functionalized with H₂S slow-releasing donors extracted from garlic, were synthetized. These innovative H₂S-releasing mats were characterized for their morphological, thermal, mechanical, and biological properties. Their antimicrobial activity and effects on the in vitro human cardiac MSC growth, either in the presence or in the absence of oxidative stress, were here assessed. On the basis of the results here presented, these new H₂S-releasing PFM could represent promising and low-cost scaffolds or patches for biomedical applications in tissue repair.
Collapse
Affiliation(s)
- Ilaria Cacciotti
- Department of Engineering, University of Rome "Niccolò Cusano", via Don Carlo Gnocchi 3, 00166 Rome, Italy.
- Italian Interuniversity Consortium on Materials Science and Technology (INSTM), 50121 Florence, Italy.
- CIMER Center for Regenerative Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133 Rome, Italy.
| | - Matteo Ciocci
- Department of Chemical Science and Technologies, University of Rome "Tor Vergata", via della Ricerca Scientifica1, 00133 Rome, Italy.
| | - Emilia Di Giovanni
- Department of Chemical Science and Technologies, University of Rome "Tor Vergata", via della Ricerca Scientifica1, 00133 Rome, Italy.
| | - Francesca Nanni
- Italian Interuniversity Consortium on Materials Science and Technology (INSTM), 50121 Florence, Italy.
- Enterprise Engineering Department, University of Rome "Tor Vergata", via del Politecnico 1, 00133 Rome, Italy.
| | - Sonia Melino
- CIMER Center for Regenerative Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133 Rome, Italy.
- Department of Chemical Science and Technologies, University of Rome "Tor Vergata", via della Ricerca Scientifica1, 00133 Rome, Italy.
| |
Collapse
|
8
|
Wang C, Lin G, Luan Y, Ding J, Li PC, Zhao Z, Qian C, Liu G, Ju S, Teng GJ. HIF-prolyl hydroxylase 2 silencing using siRNA delivered by MRI-visible nanoparticles improves therapy efficacy of transplanted EPCs for ischemic stroke. Biomaterials 2018; 197:229-243. [PMID: 30677555 DOI: 10.1016/j.biomaterials.2018.05.053] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/23/2018] [Accepted: 05/29/2018] [Indexed: 12/22/2022]
Abstract
Endothelial progenitor cell (EPC)-based therapy has brought potential benefits to stroke patients as an important restorative therapeutics. However, its efficacy is limited by poor migration and survival ability. Here, we found out that hif-prolyl hydroxylase 2 (PHD2) silencing could enhance the migration and survival ability of EPCs which could improve the therapy efficacy for ischemic stroke. We successfully developed a siRNA delivery system, which could achieve siRNA delivery and EPCs tracking with magnetic resonance imaging (MRI) simultaneously. Besides, combining MRI and bioluminescence imaging (BLI), we successfully observed full temporal profile of EPCs dynamics in vivo. Furthermore, we found out that PHD2 silencing in EPCs elevated the expression of C-X-C chemokine receptor type 4 (CXCR4) and hypoxia-inducible factor 1α (HIF-1α), which enhanced the migration and survival ability of EPCs respectively. Significantly decreased infarct volume, functional deficits and increased fractional anisotrophy (FA) value, fiber counts were observed in the siPHD2-EPCs (EPCs transfected with siRNA targeting PHD2) group. What's more, higher level of BNDF, CD31, DCX, NeuN and MBP were also observed in the siPHD2-EPCs group. Altogether, our study provides an effective method to improve EPC-based therapy efficacy for ischemic stroke.
Collapse
Affiliation(s)
- Congxiao Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Gan Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Ying Luan
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Jie Ding
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Pei-Cheng Li
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Zhen Zhao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Cheng Qian
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China.
| |
Collapse
|
9
|
Akki R, Siracusa R, Morabito R, Remigante A, Campolo M, Errami M, La Spada G, Cuzzocrea S, Marino A. Neuronal-like differentiated SH-SY5Y cells adaptation to a mild and transient H 2 O 2 -induced oxidative stress. Cell Biochem Funct 2018; 36:56-64. [PMID: 29431194 DOI: 10.1002/cbf.3317] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/13/2017] [Accepted: 12/26/2017] [Indexed: 01/30/2023]
Abstract
Preconditioning (PC) is a cell adaptive response to oxidative stress and, with regard to neurons, can be considered as a neuroprotective strategy. The aim of the present study was to verify how neuronal-like differentiated SH-SY5Y cells adapt to a mild and transient H2 O2 -induced oxidative stress and, hence, whether may be considered as more sensitive cell model to study PC pathways. A first screening allowed to define H2 O2 concentrations for PC (10μM-50μM), applied before damage(100μM H2 O2 ). Cell viability measured 24 hours after 100μM H2 O2 -induced damage was ameliorated by 24-hour pre-exposure to low-concentration H2 O2 (10μM-30μM) with cell size as well restored. Markers for apoptosis (Bcl-2 and Bad), inflammation (iNOS), and redox system (MnSOD) were also determined, showing that, in cells pre-exposed to 10μM H2 O2 and then submitted to 100μM H2 O2 , Bcl-2 levels were higher, Bad and iNOS levels were lower than those observed in damaged cells, and MnSOD levels were unchanged. Such findings show that (1) neuronal-like differentiated SH-SY5Y cells are a suitable model to investigate PC response and more sensitive to the effect of a mild and transient H2 O2 -induced oxidative stress with respect to other neuronal cells; (2) 10μM H2 O2 -induced PC is mediated by apoptotic and inflammatory pathways, unlike antioxidant system; (3) such neuroprotective strategy and underlying signals proven in neuronal-like differentiated SH-SY5Y cells may contribute to understand in vivo PC mechanisms and to define a window for pharmacological intervention, namely, related to ischemic brain damage. SIGNIFICANCE OF THE STUDY Neuronal-like differentiated SH-SY5Y cells are a suitable model to investigate PC, an endogenous neuroprotective response to a mild and transient H2 O2 -induced oxidative stress, elicited by 24-hour exposure to very low H2 O2 concentrations and mediated by both apoptotic and inflammatory pathways. This model reflects in vivo PC mechanisms occurring after brain trauma and provides novel information about pathways and time of protection useful for an appropriate pharmacological intervention.
Collapse
Affiliation(s)
- Rachid Akki
- Department of Biology, Faculty of Science, University of Abdelmalek Essaadi, Tétouan, Morocco
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Rossana Morabito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Remigante
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Mohammed Errami
- Department of Biology, Faculty of Science, University of Abdelmalek Essaadi, Tétouan, Morocco
| | - Giuseppina La Spada
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Angela Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
10
|
Rosdah AA, Bond ST, Sivakumaran P, Hoque A, Oakhill JS, Drew BG, Delbridge LMD, Lim SY. Mdivi-1 Protects Human W8B2 + Cardiac Stem Cells from Oxidative Stress and Simulated Ischemia-Reperfusion Injury. Stem Cells Dev 2017; 26:1771-1780. [PMID: 29054138 DOI: 10.1089/scd.2017.0157] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cardiac stem cell (CSC) therapy is a promising approach to treat ischemic heart disease. However, the poor survival of transplanted stem cells in the ischemic myocardium has been a major impediment in achieving an effective cell-based therapy against myocardial infarction. Inhibiting mitochondrial fission has been shown to promote survival of several cell types. However, the role of mitochondrial morphology in survival of human CSC remains unknown. In this study, we investigated whether mitochondrial division inhibitor-1 (Mdivi-1), an inhibitor of mitochondrial fission protein dynamin-related protein-1 (Drp1), can improve survival of a novel population of human W8B2+ CSCs in hydrogen peroxide (H2O2)-induced oxidative stress and simulated ischemia-reperfusion injury models. Mdivi-1 significantly reduced H2O2-induced cell death in a dose-dependent manner. This cytoprotective effect was accompanied by an increased proportion of cells with tubular mitochondria, but independent of mitochondrial membrane potential recovery and reduction of mitochondrial superoxide production. In simulated ischemia-reperfusion injury model, Mdivi-1 given as a pretreatment or throughout ischemia-reperfusion injury significantly reduced cell death. However, the cytoprotective effect of Mdivi-1 was not observed when given at reperfusion. Moreover, the cytoprotective effect of Mdivi-1 in the simulated ischemia-reperfusion injury model was not accompanied by changes in mitochondrial morphology, mitochondrial membrane potential, or mitochondrial reactive oxygen species production. Mdivi-1 also did not affect mitochondrial bioenergetics of intact W8B2+ CSCs. Taken together, these experiments demonstrated that Mdivi-1 treatment of human W8B2+ CSCs enhances their survival and can be employed to improve therapeutic efficacy of CSCs for ischemic heart disease.
Collapse
Affiliation(s)
- Ayeshah A Rosdah
- 1 St Vincent's Institute of Medical Research , Fitzroy, Australia .,2 Department of Physiology, University of Melbourne , Melbourne, Australia .,3 Faculty of Medicine, Universitas Sriwijaya , Palembang, Indonesia
| | - Simon T Bond
- 4 Molecular Metabolism and Ageing Laboratory, Baker Heart and Diabetes Institute , Melbourne, Australia
| | | | - Ashfaqul Hoque
- 1 St Vincent's Institute of Medical Research , Fitzroy, Australia
| | - Jonathan S Oakhill
- 1 St Vincent's Institute of Medical Research , Fitzroy, Australia .,5 Mary MacKillop Institute for Health Research, Australian Catholic University , Melbourne, Australia
| | - Brian G Drew
- 4 Molecular Metabolism and Ageing Laboratory, Baker Heart and Diabetes Institute , Melbourne, Australia
| | - Lea M D Delbridge
- 2 Department of Physiology, University of Melbourne , Melbourne, Australia
| | - Shiang Y Lim
- 1 St Vincent's Institute of Medical Research , Fitzroy, Australia .,6 Department of Surgery, University of Melbourne , Melbourne, Australia
| |
Collapse
|
11
|
Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M, Toietta G. Challenges and Strategies for Improving the Regenerative Effects of Mesenchymal Stromal Cell-Based Therapies. Int J Mol Sci 2017; 18:E2087. [PMID: 28974046 PMCID: PMC5666769 DOI: 10.3390/ijms18102087] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/18/2017] [Accepted: 09/28/2017] [Indexed: 12/11/2022] Open
Abstract
Cell-based therapies have the potential to revolutionize current treatments for diseases with high prevalence and related economic and social burden. Unfortunately, clinical trials have made only modest improvements in restoring normal function to degenerating tissues. This limitation is due, at least in part, to the death of transplanted cells within a few hours after transplant due to a combination of mechanical, cellular, and host factors. In particular, mechanical stress during implantation, extracellular matrix loss upon delivery, nutrient and oxygen deprivation at the recipient site, and host inflammatory response are detrimental factors limiting long-term transplanted cell survival. The beneficial effect of cell therapy for regenerative medicine ultimately depends on the number of administered cells reaching the target tissue, their viability, and their promotion of tissue regeneration. Therefore, strategies aiming at improving viable cell engraftment are crucial for regenerative medicine. Here we review the major factors that hamper successful cell engraftment and the strategies that have been studied to enhance the beneficial effects of cell therapy. Moreover, we provide a perspective on whether mesenchymal stromal cell-derived extracellular vesicle delivery, as a cell-free regenerative approach, may circumvent current cell therapy limitations.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Martina Piccoli
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research "Città della Speranza", corso Stati Uniti 4, Padova 35127, Italy.
| | - Michela Pozzobon
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| |
Collapse
|
12
|
Farías JG, Molina VM, Carrasco RA, Zepeda AB, Figueroa E, Letelier P, Castillo RL. Antioxidant Therapeutic Strategies for Cardiovascular Conditions Associated with Oxidative Stress. Nutrients 2017; 9:nu9090966. [PMID: 28862654 PMCID: PMC5622726 DOI: 10.3390/nu9090966] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress (OS) refers to the imbalance between the generation of reactive oxygen species (ROS) and the ability to scavenge these ROS by endogenous antioxidant systems, where ROS overwhelms the antioxidant capacity. Excessive presence of ROS results in irreversible damage to cell membranes, DNA, and other cellular structures by oxidizing lipids, proteins, and nucleic acids. Oxidative stress plays a crucial role in the pathogenesis of cardiovascular diseases related to hypoxia, cardiotoxicity and ischemia-reperfusion. Here, we describe the participation of OS in the pathophysiology of cardiovascular conditions such as myocardial infarction, anthracycline cardiotoxicity and congenital heart disease. This review focuses on the different clinical events where redox factors and OS are related to cardiovascular pathophysiology, giving to support for novel pharmacological therapies such as omega 3 fatty acids, non-selective betablockers and microRNAs.
Collapse
Affiliation(s)
- Jorge G Farías
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
| | - Víctor M Molina
- Unidad de Cuidados Intensivos, Hospital de Niños Roberto del Río, Santiago 7500922, Chile.
- Unidad de Cuidados Intensivos Pediátricos, Hospital Clínico Pontificia Universidad Católica de Chile, Santiago 7500922, Chile.
| | - Rodrigo A Carrasco
- Laboratorio de Investigación Biomédica, Departamento de Medicina Interna, Hospital del Salvador, Santiago 7500922, Chile.
- Departamento de Cardiología, Clínica Alemana, Santiago 7500922, Chile.
| | - Andrea B Zepeda
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
| | - Elías Figueroa
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
- Núcleo de Investigación en Producción Alimentaria, BIOACUI, Escuela de Acuicultura, Universidad Católica de Temuco, Temuco 4780000, Chile.
| | - Pablo Letelier
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
- School of Health Sciences, Universidad Católica de Temuco, Temuco 4780000, Chile.
| | - Rodrigo L Castillo
- Laboratorio de Investigación Biomédica, Departamento de Medicina Interna, Hospital del Salvador, Santiago 7500922, Chile.
- Programa de Fisiopatología Oriente, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile.
| |
Collapse
|
13
|
Müller AS, Janjić K, Lilaj B, Edelmayer M, Agis H. Hypoxia-based strategies for regenerative dentistry—Views from the different dental fields. Arch Oral Biol 2017; 81:121-130. [DOI: 10.1016/j.archoralbio.2017.04.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/24/2017] [Accepted: 04/25/2017] [Indexed: 12/20/2022]
|
14
|
Hu Z, Hu S, Yang S, Chen M, Zhang P, Liu J, Abbott GW. Remote Liver Ischemic Preconditioning Protects against Sudden Cardiac Death via an ERK/GSK-3β-Dependent Mechanism. PLoS One 2016; 11:e0165123. [PMID: 27768739 PMCID: PMC5074543 DOI: 10.1371/journal.pone.0165123] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/06/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Preconditioning stimuli conducted in remote organs can protect the heart against subsequent ischemic injury, but effects on arrhythmogenesis and sudden cardiac death (SCD) are unclear. Here, we investigated the effect of remote liver ischemia preconditioning (RLIPC) on ischemia/reperfusion (I/R)-induced cardiac arrhythmia and sudden cardiac death (SCD) in vivo, and determined the potential role of ERK/GSK-3βsignaling. METHODS/RESULTS Male Sprague Dawley rats were randomized to sham-operated, control, or RLIPC groups. RLIPC was induced by alternating four 5-minute cycles of liver ischemia with 5-minute intermittent reperfusions. To investigate I/R-induced arrhythmogenesis, hearts in each group were subsequently subjected to 5-minute left main coronary artery ligation followed by 20-minute reperfusion. RLIPC reduced post-I/R ventricular arrhythmias, and decreased the incidence of SCD >threefold. RLIPC increased phosphorylation of cardiac ERK1/2, and GSK-3β Ser9 but not Tyr216 post-I/R injury. Inhibition of either GSK-3β (with SB216763) or ERK1/2 (with U0126) abolished RLIPC-induced antiarrhythmic activity and GSK-3β Ser9 and ERK1/2 phosphorylation, leaving GSK-3β Tyr216 phosphorylation unchanged. CONCLUSIONS RLIPC exerts a powerful antiarrhythmic effect and reduces predisposition to post-IR SCD. The underlying mechanism of RLIPC cardioprotection against I/R-induced early arrhythmogenesis may involve ERK1/2/GSK-3β Ser9-dependent pathways.
Collapse
Affiliation(s)
- Zhaoyang Hu
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sheng Hu
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Shuai Yang
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mou Chen
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Zhang
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jin Liu
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- * E-mail: (GWA); (JL)
| | - Geoffrey W. Abbott
- Bioelectricity Laboratory, Dept. of Pharmacology and Dept. of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, California, United States of America
- * E-mail: (GWA); (JL)
| |
Collapse
|
15
|
Preconditioning of Human Mesenchymal Stem Cells to Enhance Their Regulation of the Immune Response. Stem Cells Int 2016; 2016:3924858. [PMID: 27822228 PMCID: PMC5086389 DOI: 10.1155/2016/3924858] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted the attention of researchers and clinicians for their ability to differentiate into a number of cell types, participate in tissue regeneration, and repair the damaged tissues by producing various growth factors and cytokines, as well as their unique immunoprivilege in alloreactive hosts. The immunomodulatory functions of exogenous MSCs have been widely investigated in immune-mediated inflammatory diseases and transplantation research. However, a harsh environment at the site of tissue injury/inflammation with insufficient oxygen supply, abundance of reactive oxygen species, and presence of other harmful molecules that damage the adoptively transferred cells collectively lead to low survival and engraftment of the transferred cells. Preconditioning of MSCs ex vivo by hypoxia, inflammatory stimulus, or other factors/conditions prior to their use in therapy is an adaptive strategy that prepares MSCs to survive in the harsh environment and to enhance their regulatory function of the local immune responses. This review focuses on a number of approaches in preconditioning human MSCs with the goal of augmenting their capacity to regulate both innate and adaptive immune responses.
Collapse
|
16
|
Saparov A, Ogay V, Nurgozhin T, Jumabay M, Chen WCW. Preconditioning of Human Mesenchymal Stem Cells to Enhance Their Regulation of the Immune Response. Stem Cells Int 2016; 2016:3924858. [PMID: 27822228 PMCID: PMC5086389 DOI: 10.1155/2016/3924858 10.1155/2016/3924858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/28/2016] [Indexed: 03/24/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted the attention of researchers and clinicians for their ability to differentiate into a number of cell types, participate in tissue regeneration, and repair the damaged tissues by producing various growth factors and cytokines, as well as their unique immunoprivilege in alloreactive hosts. The immunomodulatory functions of exogenous MSCs have been widely investigated in immune-mediated inflammatory diseases and transplantation research. However, a harsh environment at the site of tissue injury/inflammation with insufficient oxygen supply, abundance of reactive oxygen species, and presence of other harmful molecules that damage the adoptively transferred cells collectively lead to low survival and engraftment of the transferred cells. Preconditioning of MSCs ex vivo by hypoxia, inflammatory stimulus, or other factors/conditions prior to their use in therapy is an adaptive strategy that prepares MSCs to survive in the harsh environment and to enhance their regulatory function of the local immune responses. This review focuses on a number of approaches in preconditioning human MSCs with the goal of augmenting their capacity to regulate both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Arman Saparov
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan
| | - Vyacheslav Ogay
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan
| | - Talgat Nurgozhin
- Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Medet Jumabay
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - William C. W. Chen
- Research Laboratory of Electronics and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
17
|
Kim PH, Na SS, Lee B, Kim JH, Cho JY. Stanniocalcin 2 enhances mesenchymal stem cell survival by suppressing oxidative stress. BMB Rep 2016; 48:702-7. [PMID: 26424558 PMCID: PMC4791327 DOI: 10.5483/bmbrep.2015.48.12.158] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Indexed: 01/01/2023] Open
Abstract
To overcome the disadvantages of stem cell-based cell therapy like low cell survival at the disease site, we used stanniocalcin 2 (STC2), a family of secreted glycoprotein hormones that function to inhibit apoptosis and oxidative damage and to induce proliferation. STC2 gene was transfected into two kinds of stem cells to prolong cell survival and protect the cells from the damage by oxidative stress. The stem cells expressing STC2 exhibited increased cell viability and improved cell survival as well as elevated expression of the pluripotency and self-renewal markers (Oct4 and Nanog) under sub-lethal oxidative conditions. Up-regulation of CDK2 and CDK4 and down-regulation of cell cycle inhibitors p16 and p21 were observed after the delivery of STC2. Furthermore, STC2 transduction activated pAKT and pERK 1/2 signal pathways. Taken together, the STC2 can be used to enhance cell survival and maintain long-term stemness in therapeutic use of stem cells. [BMB Reports 2015; 48(12): 702-707]
Collapse
Affiliation(s)
- Pyung-Hwan Kim
- Department of Biomedical Laboratory Science, College of Medical Science, Konyang University, Daejeon 35365, Korea; Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Sang-Su Na
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Bomnaerin Lee
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Joo-Hyun Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
18
|
Melatonin Protects Human Adipose-Derived Stem Cells from Oxidative Stress and Cell Death. Arch Plast Surg 2016; 43:237-41. [PMID: 27218020 PMCID: PMC4876151 DOI: 10.5999/aps.2016.43.3.237] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 04/04/2016] [Accepted: 04/12/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Adipose-derived stem cells (ASCs) have applications in regenerative medicine based on their therapeutic potential to repair and regenerate diseased and damaged tissue. They are commonly subject to oxidative stress during harvest and transplantation, which has detrimental effects on their subsequent viability. By functioning as an antioxidant against free radicals, melatonin may exert cytoprotective effects on ASCs. METHODS We cultured human ASCs in the presence of varying dosages of hydrogen peroxide and/or melatonin for a period of 3 hours. Cell viability and apoptosis were determined with propidium iodide and Hoechst 33342 staining under fluorescence microscopy. RESULTS Hydrogen peroxide (1-2.5 mM) treatment resulted in an incremental increase in cell death. 2 mM hydrogen peroxide was thereafter selected as the dose for co-treatment with melatonin. Melatonin alone had no adverse effects on ASCs. Co-treatment of ASCs with melatonin in the presence of hydrogen peroxide protected ASCs from cell death in a dose-dependent manner, and afforded maximal protection at 100 µM (n=4, one-way analysis of variance P<0.001). Melatonin co-treated ASCs displayed significantly fewer apoptotic cells, as demonstrated by condensed and fragmented nuclei under fluorescence microscopy. CONCLUSIONS Melatonin possesses cytoprotective properties against oxidative stress in human ASCs and might be a useful adjunct in fat grafting and cell-assisted lipotransfer.
Collapse
|
19
|
Harnessing the secretome of cardiac stem cells as therapy for ischemic heart disease. Biochem Pharmacol 2016; 113:1-11. [PMID: 26903387 DOI: 10.1016/j.bcp.2016.02.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/18/2016] [Indexed: 12/22/2022]
Abstract
Adult stem cells continue to promise opportunities to repair damaged cardiac tissue. However, precisely how adult stem cells accomplish cardiac repair, especially after ischemic damage, remains controversial. It has been postulated that the clinical benefit of adult stem cells for cardiovascular disease results from the release of cytokines and growth factors by the transplanted cells. Studies in animal models of myocardial infarction have reported that such paracrine factors released from transplanted adult stem cells contribute to improved cardiac function by several processes. These include promoting neovascularization of damaged tissue, reducing inflammation, reducing fibrosis and scar formation, as well as protecting cardiomyocytes from apoptosis. In addition, these factors might also stimulate endogenous repair by activating cardiac stem cells. Interestingly, stem cells discovered to be resident in the heart appear to be functionally superior to extra-cardiac adult stem cells when transplanted for cardiac repair and regeneration. In this review, we discuss the therapeutic potential of cardiac stem cells and how the proteins secreted from these cells might be harnessed to promote repair and regeneration of damaged cardiac tissue. We also highlight how recent controversies about the efficacy of adult stem cells in clinical trials of ischemic heart disease have not dampened enthusiasm for the application of cardiac stem cells and their paracrine factors for cardiac repair: the latter have proved superior to the mesenchymal stem cells used in most clinical trials in the past, some of which appear to have been conducted with sub-optimal rigor.
Collapse
|
20
|
Hutton DL, Grayson WL. Hypoxia Inhibits De Novo Vascular Assembly of Adipose-Derived Stromal/Stem Cell Populations, but Promotes Growth of Preformed Vessels. Tissue Eng Part A 2015; 22:161-9. [PMID: 26481655 DOI: 10.1089/ten.tea.2015.0421] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Vascularization is critical for cell survival within tissue-engineered grafts. Adipose-derived stromal/stem cells (ASCs) are widely used in tissue engineering applications as they are a clinically relevant source of stem cells and endothelial progenitor cells. ASCs have previously been shown to self-assemble into pericyte-stabilized vascular networks in normoxic (20% O2) cultures. This capacity for de novo vascular assembly may accelerate graft vascularization in vivo rather than relying solely on angiogenic ingrowth. However, oxygen depletion within large cell-seeded grafts will be rapid, and it is unclear how this worsening hypoxic environment will impact the vascular assembly of the transplanted cells. The objectives of this study were to determine whether ASC-derived vessels could grow in hypoxia and to assess whether the vessel maturity (i.e., individual cells vs. preformed vessels) influenced this hypoxic response. Utilizing an in vitro vascularization model, ASCs were encapsulated within fibrin gels and cultured in vitro for up to 6 days in either normoxia (20% O2) or hypoxia (0.2% or 2% O2). In a subsequent experiment, vessels were allowed to preform in normoxia for 6 days before an additional 6 days of either normoxia or hypoxia. Viability, vessel growth, pericyte coverage, proliferation, metabolism, and angiogenic factor expression were assessed for each experimental approach. Vessel growth was dramatically inhibited in both moderate and severe hypoxia (47% and 11% total vessel length vs. normoxia, respectively), despite maintaining high cell viability and upregulating endogenous expression of vascular endothelial growth factor in hypoxia. Bromodeoxyuridine labeling indicated significantly reduced proliferation of endothelial cells in hypoxia. In contrast, when vascular networks were allowed to preform for 6 days in normoxia, vessels not only survived but also continued to grow more in hypoxia than those maintained in normoxia. These findings demonstrate that vascular assembly and growth are tightly regulated by oxygen tension and may be differentially affected by hypoxic conditions based on the maturity of the vessels. Understanding this relationship is critical to developing effective approaches to engineer viable tissue-engineered grafts in vivo.
Collapse
Affiliation(s)
- Daphne L Hutton
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Warren L Grayson
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Department of Material Sciences and Engineering, Johns Hopkins University School of Engineering , Baltimore, Maryland
| |
Collapse
|
21
|
Feng S, Zhao Y, Xian M, Wang Q. Biological thiols-triggered hydrogen sulfide releasing microfibers for tissue engineering applications. Acta Biomater 2015; 27:205-213. [PMID: 26363376 PMCID: PMC4609630 DOI: 10.1016/j.actbio.2015.09.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 09/04/2015] [Accepted: 09/08/2015] [Indexed: 01/15/2023]
Abstract
By electrospinning of polycaprolactone (PCL) solutions containing N-(benzoylthio)benzamide (NSHD1), a H2S donor, fibrous scaffolds with hydrogen sulfide (H2S) releasing capability (H2S-fibers) are fabricated. The resultant microfibers are capable of releasing H2S upon immersion in aqueous solution containing biological thiols under physiological conditions. The H2S release peaks of H2S-fibers appeared at 2-4h, while the peak of donor alone showed at 45 min. H2S release half-lives of H2S-fibers were 10-20 times longer than that of donor alone. Furthermore, H2S-fibers can protect cells from H2O2 induced oxidative damage by significantly decreasing the production of intracellular reactive oxygen species (ROS). Finally, we investigated the H2S-fibers application as a wound dressing in vitro. Given that H2S has a broad range of physiological functions, H2S-fibers hold great potential for various biomedical applications. STATEMENT OF SIGNIFICANCE Hydrogen sulfide, as a gaseous messenger, plays a crucial role in many physiological and pathological conditions. Recent studies about functions of H2S suggests H2S-based therapy could be promising therapeutic strategy for many diseases, such as cardiovascular disease, arthritis, and inflammatory bowel disease. Although many H2S donors have been developed and applied for biomedical studies, most of H2S donors have the shortage that the H2S release is either too fast or uncontrollable, which poorly mimic the biological generation of H2S. By simply combining electrospinning technique with our designed biological thiols activated H2S donor, NSHD1, we fabricated H2S releasing microfibers (H2S-fibers). This H2S-fibers significantly prolonged the releasing time compared to H2S donor alone. By adjusting the electrospinning parameters, tunable releasing profiles can be achieved. Moreover, the H2S fibers can protect cardiac myoblasts H9c2 and fibroblast NIH 3T3 from oxidative damage and support their proliferation as cellular scaffolds. To our knowledge, this is the first report of electrospun fibers with H2S releasing capacity. We anticipate this H2S-releasing scaffold will have great potential for biomedical applications.
Collapse
Affiliation(s)
- Sheng Feng
- Department of Chemistry and Biochemistry and Nanocenter, University of South Carolina, Columbia, SC 29208, USA
| | - Yu Zhao
- Department of Chemistry, Washington State University, Pullman, WA 99164, USA
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, WA 99164, USA.
| | - Qian Wang
- Department of Chemistry and Biochemistry and Nanocenter, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
22
|
Hu X, Huang X, Yang Q, Wang L, Sun J, Zhan H, Lin J, Pu Z, Jiang J, Sun Y, Xiang M, Liu X, Xie X, Yu X, Chen Z, Tse HF, Zhang J, Wang J. Safety and efficacy of intracoronary hypoxia-preconditioned bone marrow mononuclear cell administration for acute myocardial infarction patients: The CHINA-AMI randomized controlled trial. Int J Cardiol 2015; 184:446-451. [DOI: 10.1016/j.ijcard.2015.02.084] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/14/2015] [Accepted: 02/22/2015] [Indexed: 12/21/2022]
|
23
|
Shukla L, Morrison WA, Shayan R. Adipose-derived stem cells in radiotherapy injury: a new frontier. Front Surg 2015; 2:1. [PMID: 25674565 PMCID: PMC4309196 DOI: 10.3389/fsurg.2015.00001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/01/2015] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is increasingly used to treat numerous human malignancies. In addition to the beneficial anti-cancer effects, there are a series of undesirable effects on normal host tissues surrounding the target tumor. While the early effects of radiotherapy (desquamation, erythema, and hair loss) typically resolve, the chronic effects persist as unpredictable and often troublesome sequelae of cancer treatment, long after oncological treatment has been completed. Plastic surgeons are often called upon to treat the problems subsequently arising in irradiated tissues, such as recurrent infection, impaired healing, fibrosis, contracture, and/or lymphedema. Recently, it was anecdotally noted - then validated in more robust animal and human studies - that fat grafting can ameliorate some of these chronic tissue effects. Despite the widespread usage of fat grafting, the mechanism of its action remains poorly understood. This review provides an overview of the current understanding of: (i) mechanisms of chronic radiation injury and its clinical manifestations; (ii) biological properties of fat grafts and their key constituent, adipose-derived stem cells (ADSCs); and (iii) the role of ADSCs in radiotherapy-induced soft-tissue injury.
Collapse
Affiliation(s)
- Lipi Shukla
- Regenerative Surgery Group, O'Brien Institute , Fitzroy, VIC , Australia ; Department of Plastic Surgery, St. Vincent's Hospital , Fitzroy, VIC , Australia ; Regenerative Surgery Group, Australian Catholic University and O'Brien Institute Tissue Engineering Centre (AORTEC) , Fitzroy, VIC , Australia
| | - Wayne A Morrison
- Regenerative Surgery Group, O'Brien Institute , Fitzroy, VIC , Australia ; Department of Plastic Surgery, St. Vincent's Hospital , Fitzroy, VIC , Australia ; Regenerative Surgery Group, Australian Catholic University and O'Brien Institute Tissue Engineering Centre (AORTEC) , Fitzroy, VIC , Australia ; Department of Surgery, University of Melbourne , Melbourne, VIC , Australia
| | - Ramin Shayan
- Regenerative Surgery Group, O'Brien Institute , Fitzroy, VIC , Australia ; Department of Plastic Surgery, St. Vincent's Hospital , Fitzroy, VIC , Australia ; Regenerative Surgery Group, Australian Catholic University and O'Brien Institute Tissue Engineering Centre (AORTEC) , Fitzroy, VIC , Australia ; Department of Surgery, University of Melbourne , Melbourne, VIC , Australia
| |
Collapse
|
24
|
Zhang C, Guo Z, Liu H, Shi Y, Ge S. Influence of levosimendan postconditioning on apoptosis of rat lung cells in a model of ischemia-reperfusion injury. PLoS One 2015; 10:e0114963. [PMID: 25608001 PMCID: PMC4301642 DOI: 10.1371/journal.pone.0114963] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 11/17/2014] [Indexed: 12/17/2022] Open
Abstract
Objective To ascertain if levosimendan postconditioning can alleviate lung ischemia–reperfusion injury (LIRI) in rats. Method One hundred rats were divided into five groups: Sham (sham), ischemia–reperfusion group (I/R group), ischemic postconditioning (IPO group), levosimendan postconditioning (Levo group) and combination postconditioning group of levosimendan and 5-Hydroxydecanoic acid (Levo+5-HD group). The apoptotic index (AI) of lung tissue cells was determined using the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Expression of active cysteine aspartate specific protease-3 ( active caspase-3), Bcl-2 and Bax in lung tissue was determined by immunohistochemical staining. The morphopathology of lung tissue was observed using light and electron microscopy. Results AI values and expression of active caspase-3, Bcl-2 and Bax of lung tissue in I/R and Levo+5-HD groups were significantly higher than those in the sham group ( P<0.05). AI values and expression of active caspase-3 and Bax were significantly lower, whereas that of Bcl-2 was higher significantly in the Levo group, compared with I/R and Levo+5-HD groups (P<0.05). Significant differences were not observed in comparisons between I/R and Levo+5-HD groups as well as IPO and Levo groups. Conclusion LIRI can be alleviated by levosimendan, which simulates an IPO protective function. A postulated lung-protective mechanism of action could involve opening of mitochondrial adenosine triphosphate-sensitive potassium channels, relieving Ca2+ overload, upregulation of expression of Bcl-2, and downregulation of expression of active caspase-3 and Bax.
Collapse
Affiliation(s)
- Chengxin Zhang
- Cardiovascular Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhixiang Guo
- Cardiovascular Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Haiyuan Liu
- Oncology Department, The He Fei Hospital affiliated with An Hui Medical University, Hefei, Anhui, China
| | - Yinglu Shi
- Department of Cerebral Surgery, The Chest Hospital of Anhui province, Hefei, Anhui, China
| | - Shenglin Ge
- Cardiovascular Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- * E-mail:
| |
Collapse
|
25
|
Veronesi E, Burns JS, Murgia A, Candini O, Rasini V, Mastrolia I, Catani F, Paolucci P, Dominici M. cGMP-compliant transportation conditions for a prompt therapeutic use of marrow mesenchymal stromal/stem cells. Methods Mol Biol 2015; 1283:109-122. [PMID: 25108453 DOI: 10.1007/7651_2014_105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
We recently described conditions for safe 18-h manufacturer-to-patient transportation of freshly harvested hBM-MSC expanded under cGMP protocols using human platelet lysate (hPL), that allowed prompt use as an advanced therapeutic medicinal product. Here we outline important considerations when comparing different transportation conditions, highlighting that although cell transportation may involve a reduction in viability, this did not undermine the ultimate bone-forming regenerative potential of the cGMP-hBM-MSC population.
Collapse
Affiliation(s)
- Elena Veronesi
- Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41100, Modena, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Hypoxia/Reoxygenation-Preconditioned Human Bone Marrow-Derived Mesenchymal Stromal Cells Rescue Ischemic Rat Cortical Neurons by Enhancing Trophic Factor Release. Mol Neurobiol 2014; 52:792-803. [DOI: 10.1007/s12035-014-8912-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 09/28/2014] [Indexed: 02/07/2023]
|
27
|
Abstract
Stem cells have emerged as promising tools for the treatment of incurable neural and heart diseases and tissue damage. However, the survival of transplanted stem cells is reported to be low, reducing their therapeutic effects. The major causes of poor survival of stem cells in vivo are linked to anoikis, potential immune rejection, and oxidative damage mediating apoptosis. This review investigates novel methods and potential molecular mechanisms for stem cell preconditioning in vitro to increase their retention after transplantation in damaged tissues. Microenvironmental preconditioning (e.g., hypoxia, heat shock, and exposure to oxidative stress), aggregate formation, and hydrogel encapsulation have been revealed as promising strategies to reduce cell apoptosis in vivo while maintaining biological functions of the cells. Moreover, this review seeks to identify methods of optimizing cell dose preparation to enhance stem cell survival and therapeutic function after transplantation.
Collapse
Affiliation(s)
- Sébastien Sart
- Hydrodynamics Laboratory , CNRS UMR7646, Ecole Polytechnique, Palaiseau, France
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| |
Collapse
|