1
|
Li M, Guo N, Yu J, Chen J, Wang Y, Cao X, Mao Y, Yan L. Beneficial effects of the Achillea millefolium green-formulated zinc nanoparticles in mice with heart failure following myocardial infarction. INORG CHEM COMMUN 2024; 169:113005. [DOI: 10.1016/j.inoche.2024.113005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
2
|
Aglan HA, Ahmed HH, Beherei HH, Abdel-Hady BM, Ekram B, Kishta MS. Generation of cardiomyocytes from stem cells cultured on nanofibrous scaffold: Experimental approach for attenuation of myocardial infarction. Tissue Cell 2024; 89:102461. [PMID: 38991272 DOI: 10.1016/j.tice.2024.102461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/04/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
The current study was constructed to fabricate polyamide based nanofibrous scaffolds (NS) and to define the most promising one for the generation of cardiomyocytes from adipose tissue derived mesenchymal stem cells (ADMSCs). This purpose was extended to assess the potentiality of the generated cardiomyocytes in relieving myocardial infarction (MI) in rats. Production and characterization of NSs were carried out. ADMSCs were cultured on NS and induced to differentiate into cardiomyocytes by specific growth factors. Molecular analysis for myocyte-specific enhancer factor 2 C (MEF2C) and alpha sarcomeric actin (α-SCA) expression was done to confirm the differentiation of ADMSCs into cardiomyocytes for further transplantation into MI induced rats. Implantation of cells in MI afflicted rats boosted heart rate, ST height and PR interval and lessened P duration, RR, QTc and QRS intervals. Also, this type of medication minified serum lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) enzymes activity as well as serum and cardiac troponin T (Tn-T) levels and upraised serum and cardiac α-SCA and cardiac connexin 43 (CX 43) levels. Microscopic feature of cardiac tissue sections of rats in the treated groups revealed great renovation in the cardiac microarchitecture. Conclusively, this attempt gains insight into a realistic strategy for recovery of MI through systemic employment of in vitro generated cardiomyocytes.
Collapse
Affiliation(s)
- Hadeer A Aglan
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt.
| | - Hanaa H Ahmed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, Advanced Materials Technology and Mineral Resources Research Institute, National Research Centre, Giza, Egypt
| | - Bothaina M Abdel-Hady
- Polymers and Pigments Department, Chemical Industries Institute, National Research Centre, Giza, Egypt
| | - Basma Ekram
- Polymers and Pigments Department, Chemical Industries Institute, National Research Centre, Giza, Egypt
| | - Mohamed S Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| |
Collapse
|
3
|
Guan H, Chen Y, Liu X, Huang L. Research and application of hydrogel-encapsulated mesenchymal stem cells in the treatment of myocardial infarction. Colloids Surf B Biointerfaces 2024; 239:113942. [PMID: 38729022 DOI: 10.1016/j.colsurfb.2024.113942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Myocardial infarction (MI) stands out as a highly lethal disease that poses a significant threat to global health. Worldwide, heart failure resulting from MI remains a leading cause of human mortality. Mesenchymal stem cell (MSC) therapy has emerged as a promising therapeutic approach, leveraging its intrinsic healing properties. Nevertheless, pervasive issues, including a low cell retention rate, suboptimal survival rate, and incomplete differentiation of MSCs, present formidable challenges for further research. The introduction and advancement of biomaterials have offered a novel avenue for the exploration of MSC therapy in MI, marking considerable progress thus far. Notably, hydrogels, among the representative biomaterials, have garnered extensive attention within the biomedical field. This review delves into recent advancements, specifically focusing on the application of hydrogels to augment MSC therapy for cardiac tissue regeneration in MI.
Collapse
Affiliation(s)
- Haien Guan
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou 525200, China
| | - Yuehua Chen
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou 525200, China
| | - Xuanyu Liu
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou 525200, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou 525200, China.
| |
Collapse
|
4
|
Motta I, Soccio M, Guidotti G, Lotti N, Pasquinelli G. Hydrogels for Cardio and Vascular Tissue Repair and Regeneration. Gels 2024; 10:196. [PMID: 38534614 DOI: 10.3390/gels10030196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Cardiovascular disease (CVD), the leading cause of death globally, affects the heart and arteries with a variety of clinical manifestations, the most dramatic of which are myocardial infarction (MI), abdominal aortic aneurysm (AAA), and intracranial aneurysm (IA) rupture. In MI, necrosis of the myocardium, scar formation, and loss of cardiomyocytes result from insufficient blood supply due to coronary artery occlusion. Beyond stenosis, the arteries that are structurally and functionally connected to the cardiac tissue can undergo pathological dilation, i.e., aneurysmal dilation, with high risk of rupture. Aneurysms of the intracranial arteries (IAs) are more commonly seen in young adults, whereas those of the abdominal aorta (AAA) are predominantly seen in the elderly. IAs, unpredictably, can undergo rupture and cause life-threatening hemorrhage, while AAAs can result in rupture, internal bleeding and high mortality rate. In this clinical context, hydrogels, three-dimensional networks of water-seizing polymers, have emerged as promising biomaterials for cardiovascular tissue repair or protection due to their biocompatibility, tunable properties, and ability to encapsulate and release bioactive molecules. This review provides an overview of the current state of research on the use of hydrogels as an innovative platform to promote cardiovascular-specific tissue repair in MI and functional recovery or protection in aneurysmal dilation.
Collapse
Affiliation(s)
- Ilenia Motta
- Alma Mater Institute on Healthy Planet, University of Bologna, Via Massarenti 11, 40138 Bologna, Italy
| | - Michelina Soccio
- Civil, Chemical, Environmental and Materials Engineering Department, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Giulia Guidotti
- Civil, Chemical, Environmental and Materials Engineering Department, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Nadia Lotti
- Civil, Chemical, Environmental and Materials Engineering Department, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Gianandrea Pasquinelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
5
|
Gao H, Liu S, Qin S, Yang J, Yue T, Ye B, Tang Y, Feng J, Hou J, Danzeng D. Injectable hydrogel-based combination therapy for myocardial infarction: a systematic review and Meta-analysis of preclinical trials. BMC Cardiovasc Disord 2024; 24:119. [PMID: 38383333 PMCID: PMC10882925 DOI: 10.1186/s12872-024-03742-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION This study evaluates the effectiveness of a combined regimen involving injectable hydrogels for the treatment of experimental myocardial infarction. PATIENT CONCERNS Myocardial infarction is an acute illness that negatively affects quality of life and increases mortality rates. Experimental models of myocardial infarction can aid in disease research by allowing for the development of therapies that effectively manage disease progression and promote tissue repair. DIAGNOSIS Experimental animal models of myocardial infarction were established using the ligation method on the anterior descending branch of the left coronary artery (LAD). INTERVENTIONS The efficacy of intracardiac injection of hydrogels, combined with cells, drugs, cytokines, extracellular vesicles, or nucleic acid therapies, was evaluated to assess the functional and morphological improvements in the post-infarction heart achieved through the combined hydrogel regimen. OUTCOMES A literature review was conducted using PubMed, Web of Science, Scopus, and Cochrane databases. A total of 83 papers, including studies on 1332 experimental animals (rats, mice, rabbits, sheep, and pigs), were included in the meta-analysis based on the inclusion and exclusion criteria. The overall effect size observed in the group receiving combined hydrogel therapy, compared to the group receiving hydrogel treatment alone, resulted in an ejection fraction (EF) improvement of 8.87% [95% confidence interval (CI): 7.53, 10.21] and a fractional shortening (FS) improvement of 6.31% [95% CI: 5.94, 6.67] in rat models, while in mice models, the improvements were 16.45% [95% CI: 11.29, 21.61] for EF and 5.68% [95% CI: 5.15, 6.22] for FS. The most significant improvements in EF (rats: MD = 9.63% [95% CI: 4.02, 15.23]; mice: MD = 23.93% [95% CI: 17.52, 30.84]) and FS (rats: MD = 8.55% [95% CI: 2.54, 14.56]; mice: MD = 5.68% [95% CI: 5.15, 6.22]) were observed when extracellular vesicle therapy was used. Although there have been significant results in large animal experiments, the number of studies conducted in this area is limited. CONCLUSION The present study demonstrates that combining hydrogel with other therapies effectively improves heart function and morphology. Further preclinical research using large animal models is necessary for additional study and validation.
Collapse
Affiliation(s)
- Han Gao
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Song Liu
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Shanshan Qin
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Jiali Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Tian Yue
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Bengui Ye
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Yue Tang
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Feng
- School of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jun Hou
- Department of Cardiology, Chengdu Third People's Hospital, Chengdu, Sichuan, China.
| | - Dunzhu Danzeng
- School of Medicine, Tibet University, Lhasa, Tibet, China.
| |
Collapse
|
6
|
Shaik R, Xu J, Wang Y, Hong Y, Zhang G. Fibrin-Enriched Cardiac Extracellular Matrix Hydrogel Promotes In Vitro Angiogenesis. ACS Biomater Sci Eng 2023; 9:877-888. [PMID: 36630688 PMCID: PMC10064974 DOI: 10.1021/acsbiomaterials.2c01148] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Angiogenesis is essential for cardiac repair after myocardial infarction. Promoting angiogenesis has been demonstrated as an effective approach for myocardial infarction treatment. Several different strategies for inducing myocardial angiogenesis have been explored, including exogenous delivery of angiogenic genes, proteins, microRNAs, cells, and extracellular vesicles. Various types of injectable hydrogels have been investigated for cardiac tissue repair. One of the most promising injectable hydrogels in cardiac regeneration is a cardiac extracellular matrix hydrogel that is derived from decellularized porcine myocardium. It can be delivered minimally invasively via transendocardial delivery. The safety and efficacy of cardiac extracellular matrix hydrogels have been shown in small and large animal myocardial infarction models as well as clinical trials. The main mechanisms underlying the therapeutic benefits of cardiac extracellular matrix hydrogels have been elucidated and involved in the modulation of the immune response, downregulation of pathways related to heart failure progression and fibrosis, upregulation of genes important for cardiac muscle contraction, and enhancing cardiomyocyte differentiation and maturation from stem cells. However, no potent capillary network formation induced by cardiac extracellular matrix hydrogels has been reported. In this study, we tested the feasibility of incorporating a fibrin matrix into cardiac extracellular matrix hydrogels to improve the angiogenic properties of the hydrogel. Our in vitro results demonstrate that fibrin-enriched cardiac extracellular matrix hydrogels can induce robust endothelial cell tube formation from human umbilical vein endothelial cells and promote the sprouting of human mesenchymal stem cell spheroids. The obtained information from this study is very critical toward the future in vivo evaluation of fibrin-enriched cardiac extracellular matrix hydrogels in promoting myocardial angiogenesis.
Collapse
Affiliation(s)
- Rubia Shaik
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University, State College, University Park, Pennsylvania 16801, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
7
|
Injectable Hydrogel Membrane for Guided Bone Regeneration. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010094. [PMID: 36671666 PMCID: PMC9854494 DOI: 10.3390/bioengineering10010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023]
Abstract
In recent years, multicomponent hydrogels such as interpenetrating polymer networks (IPNs) have emerged as innovative biomaterials due to the synergistic combination of the properties of each network. We hypothesized that an innovative non-animal IPN hydrogel combining self-setting silanized hydroxypropyl methylcellulose (Si-HPMC) with photochemically cross-linkable dextran methacrylate (DexMA) could be a valid alternative to porcine collagen membranes in guided bone regeneration. Calvaria critical-size defects in rabbits were filled with synthetic biphasic calcium phosphate granules in conjunction with Si-HPMC; DexMA; or Si-HPMC/DexMA experimental membranes; and in a control group with a porcine collagen membrane. The synergistic effect obtained by interpenetration of the two polymer networks improved the physicochemical properties, and the gel point under visible light was reached instantaneously. Neutral red staining of murine L929 fibroblasts confirmed the cytocompatibility of the IPN. At 8 weeks, the photo-crosslinked membranes induced a similar degree of mineral deposition in the calvaria defects compared to the positive control, with 30.5 ± 5.2% for the IPN and 34.3 ± 8.2% for the collagen membrane. The barrier effect appeared to be similar in the IPN test group compared with the collagen membrane. In conclusion, this novel, easy-to-handle and apply, photochemically cross-linkable IPN hydrogel is an excellent non-animal alternative to porcine collagen membrane in guided bone regeneration procedures.
Collapse
|
8
|
Li D, Son Y, Jang M, Wang S, Zhu W. Nanoparticle Based Cardiac Specific Drug Delivery. BIOLOGY 2023; 12:biology12010082. [PMID: 36671774 PMCID: PMC9856055 DOI: 10.3390/biology12010082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Heart failure secondary to myocardial injuries is a leading cause of death worldwide. Recently, a growing number of novel therapies have emerged for injured myocardium repairment. However, delivering therapeutic agents specifically to the injured heart remains a significant challenge. Nanoparticles are the most commonly used vehicles for targeted drug delivery. Various nanoparticles have been synthesized to deliver drugs and other therapeutic molecules to the injured heart via passive or active targeting approaches, and their targeting specificity and therapeutic efficacies have been investigated. Here, we summarized nanoparticle-based, cardiac-specific drug delivery systems, their potency for treating heart diseases, and the mechanisms underlying these cardiac-targeting strategies. We also discussed the clinical studies that have employed nanoparticle-based cardiac-specific drug delivery.
Collapse
Affiliation(s)
- Dong Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Department of Cardiology, Dongfang Hospital, The Second Affiliated Hospital of Beijing University of Chinese Medicine, Beijing 100078, China
| | - Yura Son
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ 85281, USA
| | - Shu Wang
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
- Correspondence: (S.W.); (W.Z.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Correspondence: (S.W.); (W.Z.)
| |
Collapse
|
9
|
Montheil T, Simon M, Noël D, Mehdi A, Subra G, Echalier C. Silylated biomolecules: Versatile components for bioinks. Front Bioeng Biotechnol 2022; 10:888437. [PMID: 36304899 PMCID: PMC9592925 DOI: 10.3389/fbioe.2022.888437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Physical hydrogels prepared from natural biopolymers are the most popular components for bioinks. However, to improve the mechanical properties of the network, in particular its durability for long-lasting tissue engineering applications or its stiffness for bone/cartilage applications, covalent chemical hydrogels have to be considered. For that purpose, biorthogonal reactions are required to allow the inclusion of living cells within the bioink reservoir before the 3D printing procedure. Interestingly, such reactions also unlock the possibility to further multifunctionalize the network, adding bioactive moieties to tune the biological properties of the resulting printed biomaterial. Surprisingly, compared to the huge number of studies disclosing novel bioink compositions, no extensive efforts have been made by the scientific community to develop new chemical reactions meeting the requirements of both cell encapsulation, chemical orthogonality and versatile enough to be applied to a wide range of molecular components, including fragile biomolecules. That could be explained by the domination of acrylate photocrosslinking in the bioprinting field. On the other hand, proceeding chemoselectively and allowing the polymerization of any type of silylated molecules, the sol-gel inorganic polymerization was used as a crosslinking reaction to prepare hydrogels. Recent development of this strategy includes the optimization of biocompatible catalytic conditions and the silylation of highly attractive biomolecules such as amino acids, bioactive peptides, proteins and oligosaccharides. When one combines the simplicity and the versatility of the process, with the ease of functionalization of any type of relevant silylated molecules that can be combined in an infinite manner, it was obvious that a family of bioinks could emerge quickly. This review presents the sol-gel process in biocompatible conditions and the various classes of relevant silylated molecules that can be used as bioink components. The preparation of hydrogels and the kinetic considerations of the sol-gel chemistry which at least allowed cell encapsulation and extrusion-based bioprinting are discussed.
Collapse
Affiliation(s)
- Titouan Montheil
- IBMM, University Montpellier, CNRS, ENSCM, Montpellier, France
- ICGM, University Montpellier, CNRS, ENSCM, Montpellier, France
| | - Matthieu Simon
- IBMM, University Montpellier, CNRS, ENSCM, Montpellier, France
- IRMB, University Montpellier, INSERM, CHU, Montpellier, France
| | - Danièle Noël
- IRMB, University Montpellier, INSERM, CHU, Montpellier, France
| | - Ahmad Mehdi
- ICGM, University Montpellier, CNRS, ENSCM, Montpellier, France
| | - Gilles Subra
- IBMM, University Montpellier, CNRS, ENSCM, Montpellier, France
| | - Cécile Echalier
- IBMM, University Montpellier, CNRS, ENSCM, Montpellier, France
| |
Collapse
|
10
|
Kerignard E, Bethry A, Falcoz C, Nottelet B, Pinese C. Design of Hybrid Polymer Nanofiber/Collagen Patches Releasing IGF and HGF to Promote Cardiac Regeneration. Pharmaceutics 2022; 14:1854. [PMID: 36145603 PMCID: PMC9502465 DOI: 10.3390/pharmaceutics14091854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death globally. Myocardial infarction in particular leads to a high rate of mortality, and in the case of survival, to a loss of myocardial functionality due to post-infarction necrosis. This functionality can be restored by cell therapy or biomaterial implantation, and the need for a rapid regeneration has led to the development of bioactive patches, in particular through the incorporation of growth factors (GF). In this work, we designed hybrid patches composed of polymer nanofibers loaded with HGF and IGF and associated with a collagen membrane. Among the different copolymers studied, the polymers and their porogens PLA-Pluronic-PLA + PEG and PCL + Pluronic were selected to encapsulate HGF and IGF. While 89 and 92% of IGF were released in 2 days, HGF was released up to 58% and 50% in 35 days from PLA-Pluronic-PLA + PEG and PCL + Pluronic nanofibers, respectively. We also compared two ways of association for the loaded nanofibers and the collagen membrane, namely a direct deposition of the nanofibers on a moisturized collagen membrane (wet association), or entrapment between collagen layers (sandwich association). The interfacial cohesion and the degradation properties of the patches were evaluated. We also show that the sandwich association decreases the burst release of HGF while increasing the release efficiency. Finally, we show that the patches are cytocompatible and that the presence of collagen and IGF promotes the proliferation of C2C12 myoblast cells for 11 days. Taken together, these results show that these hybrid patches are of interest for cardiac muscle regeneration.
Collapse
|
11
|
Guimarães VY, Zanoni DS, Alves CEF, Amorim RL, Takahira RK. Immunohematological features of free-living Alouatta belzebul (Linnaeus, 1766) red-handed howler monkeys in the Eastern Amazon. Primates 2022; 63:671-682. [PMID: 35972703 DOI: 10.1007/s10329-022-01009-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 08/03/2022] [Indexed: 11/30/2022]
Abstract
The red-handed howler monkey (Alouatta belzebul) is one of the 35 threatened Brazilian primate species found in two highly endangered Brazilian biomes. Their Amazonian native populations have been declining due to exponential deforestation associated with human activities, especially the construction of dams. The studied population (n = 27) was located in the Belo Monte dam Area of Influence. For the first time, we presented hematological parameters and the basic profile of T (CD3) and B (BSAP PAX5) cells by immunocytochemistry. The results supported the hypothesis that the immuno-hematological profile is influenced by sex, age, and season. Eosinophils were significantly higher in females (p = 0.03), monocytes statistically greater in juveniles (p = 0.04), and total plasma protein increased significantly (p > 0.001) during the dry season. Furthermore, adults showed a statistically higher average absolute number of B lymphocytes than young individuals (p = 0.03), in contrast to T lymphocytes. Even without knowing the full history of antigenic exposure, these results not only contribute to elucidating the boundaries between health and disease but may help lay the groundwork for future research into the effects of anthropogenic stress on immune activation.
Collapse
Affiliation(s)
- Victor Yunes Guimarães
- Veterinary Clinical Laboratory, Veterinary Clinic Department, School of Veterinary Medicine and Animal Science, São Paulo State University, Prof. Doutor Walter Mauricio Correa St., Botucatu, SP, 18618-681, Brazil.
| | - Diogo Sousa Zanoni
- Veterinary Clinic Department, School of Veterinary Medicine and Animal Science, São Paulo State University, Prof. Doutor Walter Mauricio Correa St., Botucatu, SP, 18618-681, Brazil
| | | | - Reneé Laufer Amorim
- Veterinary Clinic Department, School of Veterinary Medicine and Animal Science, São Paulo State University, Prof. Doutor Walter Mauricio Correa St., Botucatu, SP, 18618-681, Brazil
| | - Regina Kiomi Takahira
- Veterinary Clinical Laboratory, Veterinary Clinic Department, School of Veterinary Medicine and Animal Science, São Paulo State University, Prof. Doutor Walter Mauricio Correa St., Botucatu, SP, 18618-681, Brazil
| |
Collapse
|
12
|
Demarquay C, Moussa L, Réthoré G, Milliat F, Weiss P, Mathieu N. Embedding MSCs in Si-HPMC hydrogel decreased MSC-directed host immune response and increased the regenerative potential of macrophages. Regen Biomater 2022; 9:rbac022. [PMID: 35784096 PMCID: PMC9245650 DOI: 10.1093/rb/rbac022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/22/2022] [Accepted: 04/10/2022] [Indexed: 11/14/2022] Open
Abstract
Embedding mesenchymal stromal cells (MSCs) in biomaterial is a subject of increasing interest in the field of Regenerative Medicine. Speeding up the clinical use of MSCs is dependent on the use of non-syngeneic models in accordance with Good Manufacturing Practices (GMP) requirements and on costs. To this end, in this study, we analyzed the in vivo host immune response following local injection of silanized hydroxypropyl methylcellulose (Si-HPMC)-embedded human MSCs in a rat model developing colorectal damage induced by ionizing radiation. Plasma and lymphocytes from mesenteric lymph nodes were harvested in addition to colonic tissue. We set up tests, using flow cytometry and a live imaging system, to highlight the response to specific antibodies and measure the cytotoxicity of lymphocytes against injected MSCs. We demonstrated that Si-HPMC protects MSCs from specific antibodies production and from apoptosis by lymphocytes. We also observed that Si-HPMC does not modify innate immune response infiltrate in vivo, and that in vitro co-culture of Si-HPMC-embedded MSCs impacts macrophage inflammatory response depending on the microenvironment but, more importantly, increases the macrophage regenerative response through Wnt-family and VEGF gene expression. This study furthers our understanding of the mechanisms involved, with a view to improving the therapeutic benefits of biomaterial-assisted cell therapy by modulating the host immune response. The decrease in specific immune response against injected MSCs protected by Si-HPMC also opens up new possibilities for allogeneic clinical use.
Collapse
Affiliation(s)
- Christelle Demarquay
- Human Health Department, IRSN, French Institute for Radiological Protection and Nuclear Safety, SERAMED, LRMed, Fontenay-aux-Roses 92262, France
| | - Lara Moussa
- Human Health Department, IRSN, French Institute for Radiological Protection and Nuclear Safety, SERAMED, LRMed, Fontenay-aux-Roses 92262, France
| | - Gildas Réthoré
- Faculté de Chirurgie Dentaire, Regenerative Medicine and Skeleton (RMeS) Laboratory, Université de Nantes, Nantes 44042, France
| | - Fabien Milliat
- Human Health Department, IRSN, French Institute for Radiological Protection and Nuclear Safety, SERAMED, LRMed, Fontenay-aux-Roses 92262, France
| | - Pierre Weiss
- Faculté de Chirurgie Dentaire, Regenerative Medicine and Skeleton (RMeS) Laboratory, Université de Nantes, Nantes 44042, France
| | | |
Collapse
|
13
|
Zena LA, Ekström A, Gräns A, Olsson C, Axelsson M, Sundh H, Sandblom E. It takes time to heal a broken heart: ventricular plasticity improves heart performance after myocardial infarction in rainbow trout, Oncorhynchus mykiss. J Exp Biol 2021; 224:273477. [PMID: 34792140 DOI: 10.1242/jeb.243578] [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] [Received: 09/22/2021] [Accepted: 11/12/2021] [Indexed: 12/28/2022]
Abstract
Coronary arteriosclerosis is a common feature of both wild and farmed salmonid fishes and may be linked to stress-induced cardiac pathologies. Yet, the plasticity and capacity for long-term myocardial restructuring and recovery following a restriction in coronary blood supply are unknown. Here, we analyzed the consequences of acute (3 days) and chronic (from 33 to 62 days) coronary occlusion (i.e. coronary artery ligation) on cardiac morphological characteristics and in vivo function in juvenile rainbow trout, Oncorhynchus mykiss. Acute coronary artery occlusion resulted in elevated resting heart rate and decreased inter-beat variability, which are both markers of autonomic dysfunction following acute myocardial ischemia, along with severely reduced heart rate scope (maximum-resting heart rate) relative to sham-operated trout. We also observed a loss of myocardial interstitial collagen and compact myocardium. Following long-term coronary artery ligation, resting heart rate and heart rate scope normalized relative to sham-operated trout. Moreover, a distinct fibrous collagen layer separating the compact myocardium into two layers had formed. This may contribute to maintain ventricular integrity across the cardiac cycle or, alternatively, demark a region of the compact myocardium that continues to receive oxygen from the luminal venous blood. Taken together, we demonstrate that rainbow trout may cope with the aversive effects caused by coronary artery obstruction through plastic ventricular remodeling, which, at least in part, restores cardiac performance and myocardium oxygenation.
Collapse
Affiliation(s)
- Lucas A Zena
- Department of Physiology, Institute of Biosciences, University of São Paulo, 05508-090 São Paulo, Brazil.,Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Andreas Ekström
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Albin Gräns
- Department of Animal Environment and Health, Swedish University of Agricultural Sciences, Gothenburg 405 30, Sweden
| | - Catharina Olsson
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Michael Axelsson
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Henrik Sundh
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Erik Sandblom
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden
| |
Collapse
|
14
|
Recent Advances in Cardiac Tissue Engineering for the Management of Myocardium Infarction. Cells 2021; 10:cells10102538. [PMID: 34685518 PMCID: PMC8533887 DOI: 10.3390/cells10102538] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022] Open
Abstract
Myocardium Infarction (MI) is one of the foremost cardiovascular diseases (CVDs) causing death worldwide, and its case numbers are expected to continuously increase in the coming years. Pharmacological interventions have not been at the forefront in ameliorating MI-related morbidity and mortality. Stem cell-based tissue engineering approaches have been extensively explored for their regenerative potential in the infarcted myocardium. Recent studies on microfluidic devices employing stem cells under laboratory set-up have revealed meticulous events pertaining to the pathophysiology of MI occurring at the infarcted site. This discovery also underpins the appropriate conditions in the niche for differentiating stem cells into mature cardiomyocyte-like cells and leads to engineering of the scaffold via mimicking of native cardiac physiological conditions. However, the mode of stem cell-loaded engineered scaffolds delivered to the site of infarction is still a challenging mission, and yet to be translated to the clinical setting. In this review, we have elucidated the various strategies developed using a hydrogel-based system both as encapsulated stem cells and as biocompatible patches loaded with cells and applied at the site of infarction.
Collapse
|
15
|
Bioactive Scaffolds in Stem Cell-Based Therapies for Myocardial Infarction: a Systematic Review and Meta-Analysis of Preclinical Trials. Stem Cell Rev Rep 2021; 18:2104-2136. [PMID: 34463903 DOI: 10.1007/s12015-021-10186-y] [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] [Accepted: 05/12/2021] [Indexed: 10/20/2022]
Abstract
The use of bioactive scaffolds in conjunction with stem cell therapies for cardiac repair after a myocardial infarction shows significant promise for clinical translation. We performed a systematic review and meta-analysis of preclinical trials that investigated the use of bioactive scaffolds to support stem cell-aided cardiac regeneration, in comparison to stem cell treatment alone. Cochrane Library, Medline, Embase, PubMed, Scopus, Web of Science, and grey literature were searched through April 23, 2020 and 60 articles were included in the final analysis. The overall effect size observed in scaffold and stem cell-treated small animals compared to stem cell-treated controls for ejection fraction (EF) was 7.98 [95% confidence interval (CI): 6.36, 9.59] and for fractional shortening (FS) was 5.50 [95% CI: 4.35, 6.65] in small animal models. The largest improvements in EF and FS were observed when hydrogels were used (MD = 8.45 [95% CI: 6.46, 10.45] and MD = 5.76 [95% CI: 4.46, 7.05], respectively). Subgroup analysis revealed that cardiac progenitor cells had the largest effect size for FS, and was significant from pluripotent, mesenchymal and endothelial stem cell types. In large animal studies, the overall improvement of EF favoured the use of stem cell-embedded scaffolds compared to direct injection of cells (MD = 10.49 [95% CI: 6.30, 14.67]). Significant publication bias was present in the small animal trials for EF and FS. This study supports the use of bioactive scaffolds to aid in stem cell-based cardiac regeneration. Hydrogels should be further investigated in larger animal models for clinical translation.
Collapse
|
16
|
Hoeeg C, Dolatshahi-Pirouz A, Follin B. Injectable Hydrogels for Improving Cardiac Cell Therapy-In Vivo Evidence and Translational Challenges. Gels 2021; 7:gels7010007. [PMID: 33499287 PMCID: PMC7859914 DOI: 10.3390/gels7010007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Cell therapy has the potential to regenerate cardiac tissue and treat a variety of cardiac diseases which are currently without effective treatment. This novel approach to treatment has demonstrated clinical efficiency, despite low retention of the cell products in the heart. It has been shown that improving retention often leads to improved functional outcome. A feasible method of improving cell graft retention is administration of injectable hydrogels. Over the last decade, a variety of injectable hydrogels have been investigated preclinically for their potential to improve the effects of cardiac cell therapy. These hydrogels are created with different polymers, properties, and additional functional motifs and differ in their approaches for encapsulating different cell types. Only one combinational therapy has been tested in a clinical randomized controlled trial. In this review, the latest research on the potential of injectable hydrogels for delivery of cell therapy is discussed, together with potential roadblocks for clinical translation and recommendations for future explorations to facilitate future translation.
Collapse
Affiliation(s)
- Cecilie Hoeeg
- Cardiology Stem Cell Centre, Rigshospitalet, Henrik Harpestrengs Vej 4C, 2100 Copenhagen, Denmark;
| | - Alireza Dolatshahi-Pirouz
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kongens Lyngby, Denmark;
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Dentistry—Regenerative Biomaterials, Philips van Leydenlaan 25, 6525EX Nijmegen, The Netherlands
| | - Bjarke Follin
- Cardiology Stem Cell Centre, Rigshospitalet, Henrik Harpestrengs Vej 4C, 2100 Copenhagen, Denmark;
- Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
- Correspondence:
| |
Collapse
|
17
|
Riaud M, Martinez MC, Montero-Menei CN. Scaffolds and Extracellular Vesicles as a Promising Approach for Cardiac Regeneration after Myocardial Infarction. Pharmaceutics 2020; 12:E1195. [PMID: 33317141 PMCID: PMC7763019 DOI: 10.3390/pharmaceutics12121195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/14/2022] Open
Abstract
Clinical studies have demonstrated the regenerative potential of stem cells for cardiac repair over the past decades, but their widespread use is limited by the poor tissue integration and survival obtained. Natural or synthetic hydrogels or microcarriers, used as cell carriers, contribute to resolving, in part, the problems encountered by providing mechanical support for the cells allowing cell retention, survival and tissue integration. Moreover, hydrogels alone also possess mechanical protective properties for the ischemic heart. The combined effect of growth factors with cells and an appropriate scaffold allow a therapeutic effect on myocardial repair. Despite this, the effects obtained with cell therapy remain limited and seem to be equivalent to the effects obtained with extracellular vesicles, key actors in intercellular communication. Extracellular vesicles have cardioprotective effects which, when combined proangiogenic properties with antiapoptotic and anti-inflammatory actions, make it possible to act on all the damages caused by ischemia. The evolution of biomaterial engineering allows us to envisage their association with new major players in cardiac therapy, extracellular vesicles, in order to limit undesirable effects and to envisage a transfer to the clinic. This new therapeutic approach could be associated with the release of growth factors to potentialized the beneficial effect obtained.
Collapse
Affiliation(s)
- Melody Riaud
- SOPAM, U1063, INSERM, UNIV Angers, SFR ICAT, F-49800 Angers, France;
- CRCINA, UMR 1232, INSERM, Université de Nantes, Université d’Angers, F-49933 Angers, France
| | | | | |
Collapse
|
18
|
Flegeau K, Toquet C, Rethore G, d'Arros C, Messager L, Halgand B, Dupont D, Autrusseau F, Lesoeur J, Veziers J, Bordat P, Bresin A, Guicheux J, Delplace V, Gautier H, Weiss P. In Situ Forming, Silanized Hyaluronic Acid Hydrogels with Fine Control Over Mechanical Properties and In Vivo Degradation for Tissue Engineering Applications. Adv Healthc Mater 2020; 9:e2000981. [PMID: 32864869 DOI: 10.1002/adhm.202000981] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/09/2020] [Indexed: 12/19/2022]
Abstract
In situ forming hydrogels that can be injected into tissues in a minimally-invasive fashion are appealing as delivery vehicles for tissue engineering applications. Ideally, these hydrogels should have mechanical properties matching those of the host tissue, and a rate of degradation adapted for neo-tissue formation. Here, the development of in situ forming hyaluronic acid hydrogels based on the pH-triggered condensation of silicon alkoxide precursors into siloxanes is reported. Upon solubilization and pH adjustment, the low-viscosity precursor solutions are easily injectable through fine-gauge needles prior to in situ gelation. Tunable mechanical properties (stiffness from 1 to 40 kPa) and associated tunable degradability (from 4 days to more than 3 weeks in vivo) are obtained by varying the degree of silanization (from 4.3% to 57.7%) and molecular weight (120 and 267 kDa) of the hyaluronic acid component. Following cell encapsulation, high cell viability (> 80%) is obtained for at least 7 days. Finally, the in vivo biocompatibility of silanized hyaluronic acid gels is verified in a subcutaneous mouse model and a relationship between the inflammatory response and the crosslink density is observed. Silanized hyaluronic acid hydrogels constitute a tunable hydrogel platform for material-assisted cell therapies and tissue engineering applications.
Collapse
Affiliation(s)
- Killian Flegeau
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- HTL S.A.S 7 Rue Alfred Kastler Javené 35133 France
| | - Claire Toquet
- Department of Pathology University Hospital of Nantes Nantes F‐44042 France
| | - Gildas Rethore
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| | - Cyril d'Arros
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
| | - Léa Messager
- HTL S.A.S 7 Rue Alfred Kastler Javené 35133 France
| | - Boris Halgand
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| | - Davy Dupont
- HTL S.A.S 7 Rue Alfred Kastler Javené 35133 France
| | - Florent Autrusseau
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
| | - Julie Lesoeur
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- SC3M SFR Santé F. Bonamy FED 4203 UMS Inserm 016 CNRS 3556 Nantes F‐44042 France
| | - Joëlle Veziers
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
- SC3M SFR Santé F. Bonamy FED 4203 UMS Inserm 016 CNRS 3556 Nantes F‐44042 France
| | | | | | - Jérôme Guicheux
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| | - Vianney Delplace
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
| | - Hélène Gautier
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- Université de Nantes Faculté de Pharmacie Laboratoire de Pharmacie Galénique Nantes F‐44042 France
| | - Pierre Weiss
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| |
Collapse
|
19
|
Li C, Naveed M, Dar K, Liu Z, Baig MMFA, Lv R, Saeed M, Dingding C, Feng Y, Xiaohui Z. Therapeutic advances in cardiac targeted drug delivery: from theory to practice. J Drug Target 2020; 29:235-248. [PMID: 32933319 DOI: 10.1080/1061186x.2020.1818761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The most commonly used administration methods in clinics and life are oral administration, intravenous injection, and other systemic administration methods. Targeted administration must be an essential long-term development direction due to the limited availability and a high incidence of systemic side effects. Cardiovascular diseases (CVD) are the leading cause of death all over the world. Targeted drug delivery (TDD) methods with the heart as the target organ have developed rapidly and are diversified. This article reviews the research progress of various TDD methods around the world with a heart as the target organ. It is mainly divided into two parts: the targeting vector represented by nanoparticles and various TDD methods such as intracoronary injection, ventricular wall injection, pericardial injection, and implantable medical device therapy and put forward some suggestions on the development of targeting. Different TDD methods described in this paper have not been widely used in clinical practice, and some have not even completed preclinical studies. Targeted drug delivery still requires long-term efforts by many researchers to realize the true meaning of the heart. HIGHLIGHTS Targeted administration can achieve a better therapeutic effect and effectively reduce the occurrence of adverse reactions. Parenteral administration or medical device implantation can be used for targeted drug delivery. Combined with new dosage forms or new technologies, better-targeted therapy can be achieved. Clinical trials have confirmed the safety and effectiveness of several administration methods.
Collapse
Affiliation(s)
- Cuican Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Muhammad Naveed
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China.,School of Pharmacy, Nanjing Medical University, Nanjing, P. R. China
| | - Kashif Dar
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, P. R. China
| | - Ziwei Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, P. R. China
| | - Rundong Lv
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Muhammad Saeed
- Faculty of Animal Production and Technology, The Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Chen Dingding
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Yu Feng
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Zhou Xiaohui
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China.,Department of Heart Surgery, Nanjing Shuiximen Hospital, Nanjing, P. R. China.,Department of Cardiothoracic Surgery, Zhongda Hospital affiliated with Southeast University, Nanjing, P. R. China
| |
Collapse
|
20
|
Quantifying Oxygen Levels in 3D Bioprinted Cell-Laden Thick Constructs with Perfusable Microchannel Networks. Polymers (Basel) 2020; 12:polym12061260. [PMID: 32486307 PMCID: PMC7361700 DOI: 10.3390/polym12061260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 11/28/2022] Open
Abstract
The survival and function of thick tissue engineered implanted constructs depends on pre-existing, embedded, functional, vascular-like structures that are able to integrate with the host vasculature. Bioprinting was employed to build perfusable vascular-like networks within thick constructs. However, the improvement of oxygen transportation facilitated by these vascular-like networks was directly quantified. Using an optical fiber oxygen sensor, we measured the oxygen content at different positions within 3D bioprinted constructs with and without perfusable microchannel networks. Perfusion was found to play an essential role in maintaining relatively high oxygen content in cell-laden constructs and, consequently, high cell viability. The concentration of oxygen changes following switching on and off the perfusion. Oxygen concentration depletes quickly after pausing perfusion but recovers rapidly after resuming the perfusion. The quantification of oxygen levels within cell-laden hydrogel constructs could provide insight into channel network design and cellular responses.
Collapse
|
21
|
Audam TN, Nong Y, Tomlin A, Jurkovic A, Li H, Zhu X, Long BW, Zheng YW, Weirick T, Brittian KR, Riggs DW, Gumpert A, Uchida S, Guo Y, Wysoczynski M, Jones SP. Cardiac mesenchymal cells from failing and nonfailing hearts limit ventricular dilation when administered late after infarction. Am J Physiol Heart Circ Physiol 2020; 319:H109-H122. [PMID: 32442025 DOI: 10.1152/ajpheart.00114.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although cell therapy-mediated cardiac repair offers promise for treatment/management of heart failure, lack of fundamental understanding of how cell therapy works limits its translational potential. In particular, whether reparative cells from failing hearts differ from cells derived from nonfailing hearts remains unexplored. Here, we assessed differences between cardiac mesenchymal cells (CMC) derived from failing (HF) versus nonfailing (Sham) hearts and whether the source of donor cells (i.e., from HF vs. Sham) limits reparative capacity, particularly when administered late after infarction. To determine the impact of the donor source of CMCs, we characterized the transcriptional profile of CMCs isolated from sham (Sham-CMC) and failing (HF-CMC) hearts. RNA-seq analysis revealed unique transcriptional signatures in Sham-CMC and HF-CMC, suggesting that the donor source impacts CMC. To determine whether the donor source affects reparative potential, C57BL6/J female mice were subjected to 60 min of regional myocardial ischemia and then reperfused for 35 days. In a randomized, controlled, and blinded fashion, vehicle, HF-CMC, or Sham-CMC were injected into the lumen of the left ventricle at 35 days post-MI. An additional 5 weeks later, cardiac function was assessed by echocardiography, which indicated that delayed administration of Sham-CMC and HF-CMC attenuated ventricular dilation. We also determined whether Sham-CMC and HF-CMC treatments affected ventricular histopathology. Our data indicate that the donor source (nonfailing vs. failing hearts) affects certain aspects of CMC, and these insights may have implications for future studies. Our data indicate that delayed administration of CMC limits ventricular dilation and that the source of CMC may influence their reparative actions.NEW & NOTEWORTHY Most preclinical studies have used only cells from healthy, nonfailing hearts. Whether donor condition (i.e., heart failure) impacts cells used for cell therapy is not known. We directly tested whether donor condition impacted the reparative effects of cardiac mesenchymal cells in a chronic model of myocardial infarction. Although cells from failing hearts differed in multiple aspects, they retained the potential to limit ventricular remodeling.
Collapse
Affiliation(s)
- Timothy N Audam
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Yibing Nong
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Alex Tomlin
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Andrea Jurkovic
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Hong Li
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Xiaoping Zhu
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Bethany W Long
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Yi Wei Zheng
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Tyler Weirick
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky.,Cardiovascular Innovation Institute, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Kenneth R Brittian
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Daniel W Riggs
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Anna Gumpert
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Shizuka Uchida
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky.,Cardiovascular Innovation Institute, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Yiru Guo
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Marcin Wysoczynski
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Steven P Jones
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
22
|
Boyer C, Réthoré G, Weiss P, d’Arros C, Lesoeur J, Vinatier C, Halgand B, Geffroy O, Fusellier M, Vaillant G, Roy P, Gauthier O, Guicheux J. A Self-Setting Hydrogel of Silylated Chitosan and Cellulose for the Repair of Osteochondral Defects: From in vitro Characterization to Preclinical Evaluation in Dogs. Front Bioeng Biotechnol 2020; 8:23. [PMID: 32117912 PMCID: PMC7025592 DOI: 10.3389/fbioe.2020.00023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022] Open
Abstract
Articular cartilage (AC) may be affected by many injuries including traumatic lesions that predispose to osteoarthritis. Currently there is no efficient cure for cartilage lesions. In that respect, new strategies for regenerating AC are contemplated with interest. In this context, we aim to develop and characterize an injectable, self-hardening, mechanically reinforced hydrogel (Si-HPCH) composed of silanised hydroxypropymethyl cellulose (Si-HPMC) mixed with silanised chitosan. The in vitro cytocompatibility of Si-HPCH was tested using human adipose stromal cells (hASC). In vivo, we first mixed Si-HPCH with hASC to observe cell viability after implantation in nude mice subcutis. Si-HPCH associated or not with canine ASC (cASC), was then tested for the repair of osteochondral defects in canine femoral condyles. Our data demonstrated that Si-HPCH supports hASC viability in culture. Moreover, Si-HPCH allows the transplantation of hASC in the subcutis of nude mice while maintaining their viability and secretory activity. In the canine osteochondral defect model, while the empty defects were only partially filled with a fibrous tissue, defects filled with Si-HPCH with or without cASC, revealed a significant osteochondral regeneration. To conclude, Si-HPCH is an injectable, self-setting and cytocompatible hydrogel able to support the in vitro and in vivo viability and activity of hASC as well as the regeneration of osteochondral defects in dogs when implanted alone or with ASC.
Collapse
Affiliation(s)
- Cécile Boyer
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
| | - Gildas Réthoré
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- CHU Nantes, Service d’Odontologie Restauratrice et Chirurgicale, PHU4 OTONN, Nantes, France
| | - Pierre Weiss
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- CHU Nantes, Service d’Odontologie Restauratrice et Chirurgicale, PHU4 OTONN, Nantes, France
| | - Cyril d’Arros
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
| | - Julie Lesoeur
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- SC3M – “Electron Microscopy, Microcharacterization and Functional Morphohistology Imaging” Core Facility, Structure Fédérative de Recherche Franc̨ois Bonamy, INSERM – UMS016, CNRS 3556, CHU Nantes, Université de Nantes, Nantes, France
| | - Claire Vinatier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- SC3M – “Electron Microscopy, Microcharacterization and Functional Morphohistology Imaging” Core Facility, Structure Fédérative de Recherche Franc̨ois Bonamy, INSERM – UMS016, CNRS 3556, CHU Nantes, Université de Nantes, Nantes, France
| | - Boris Halgand
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- CHU Nantes, PHU4 OTONN, Nantes, France
| | - Olivier Geffroy
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Marion Fusellier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Gildas Vaillant
- CHU Nantes, PHU4 OTONN, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Patrice Roy
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Olivier Gauthier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Jérôme Guicheux
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- SC3M – “Electron Microscopy, Microcharacterization and Functional Morphohistology Imaging” Core Facility, Structure Fédérative de Recherche Franc̨ois Bonamy, INSERM – UMS016, CNRS 3556, CHU Nantes, Université de Nantes, Nantes, France
- CHU Nantes, PHU4 OTONN, Nantes, France
| |
Collapse
|
23
|
Affiliation(s)
- Eugene Braunwald
- From the TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
24
|
Zhang J, Xu Y, Liu T, Min J, Ma Y, Song Y, Lu J, Mi W, Wang Y, Li H, Li W, Zhao D. In vivo construction of lymphoid node by implantation of adipose-derived stromal cells with hydroxypropyl methyl cellulose hydrogel in BALB/c nude mice. Organogenesis 2019; 15:85-99. [PMID: 31448695 DOI: 10.1080/15476278.2019.1656994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Adipose-derived stromal cells have multilineage potential to differentiate into several specialized tissue types. Herein, we investigated whether ADSCs could differentiate into lymphoid node in vivo. Human ADSCs from routine liposuction were cultured in differentiation medium and were supplemented with transforming growth factor β1 (TGF)-β1 and basic fibroblast growth factor (bFGF). The induced hADSCs mixed with 13% (w/v) hydroxypropyl methylcellulose (HPMC) were injected into BALB/c nude mice subcutaneously. Eight weeks later, nodules were found under the injected sites. Histology, immunohistochemistry, and species identification analysis confirmed that the nodules were lymphoid nodes that were derived from the injected hADSCs. Our experiment demonstrated that the hADSCs could differentiate into lymphocyte-like cells and form lymphoid nodes in vivo. TGF-β1 and bFGF might play important roles in the differentiation of hADSCs into lymphocyte-like cells. Our study might present an alternative approach for engineering immune organs and thus offer potential treatment for immunodeficiency diseases.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Otolaryngology Head and Neck Surgery, the second affiliated hospital.,Department of Nephrology and Endocrinology, No.371 Central Hospital of People's Liberation Army , Xinxiang , Henan , People's Republic of China
| | - Yuqiao Xu
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Tao Liu
- Faculty of Biomedical Engineering
| | - Jie Min
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Yu Ma
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Yongli Song
- Department of Otolaryngology Head and Neck Surgery, the first affiliated hospital , Xi'an , China
| | - Jianrong Lu
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Wenjuan Mi
- Department of Otolaryngology Head and Neck Surgery, the first affiliated hospital , Xi'an , China
| | - Yingmei Wang
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Hang Li
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Wangzhou Li
- Department of Plastic and Burns, the second affiliated hospital; Air Force Medical University , Xi'an , PR China
| | - Daqing Zhao
- Department of Otolaryngology Head and Neck Surgery, the second affiliated hospital
| |
Collapse
|
25
|
Talovic M, Patel K, Schwartz M, Madsen J, Garg K. Decellularized extracellular matrix gelloids support mesenchymal stem cell growth and function in vitro. J Tissue Eng Regen Med 2019; 13:1830-1842. [PMID: 31306568 DOI: 10.1002/term.2933] [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: 02/04/2019] [Revised: 06/10/2019] [Accepted: 07/03/2019] [Indexed: 12/15/2022]
Abstract
Volumetric muscle loss (VML) injuries are irrecoverable due to a significant loss of regenerative elements, persistent inflammation, extensive fibrosis, and functional impairment. When used in isolation, previous stem cell and biomaterial-based therapies have failed to regenerate skeletal muscle at clinically relevant levels. The extracellular matrix (ECM) microenvironment is crucial for the viability, stemness, and differentiation of stem cells. Decellularized-ECM (D-ECM) scaffolds are at the forefront of ongoing research to develop a viable therapy for VML. Due to the retention of key ECM components, D-ECM scaffolds provide an excellent substrate for the adhesion and migration of several cell types. Mesenchymal stem cells (MSCs) possess regenerative and immunomodulatory properties and are currently under investigation in clinical trials for a wide range of medical conditions. However, a major limitation to the use of MSCs in clinical applications is their poor viability at the site of transplantation. In this study, we have fabricated spherical scaffolds composed of gelatin and skeletal muscle D-ECM for the adhesion and delivery of MSCs to the site of VML injury. These spherical scaffolds termed "gelloids" supported MSC survival, expansion, trophic factor secretion, immunomodulation, and myogenic protein expression in vitro. Future studies would determine the therapeutic efficacy of this approach in a murine model of VML injury.
Collapse
Affiliation(s)
- Muhamed Talovic
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| | - Krishna Patel
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| | - Mark Schwartz
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| | - Josh Madsen
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| | - Koyal Garg
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| |
Collapse
|
26
|
Moussa L, Demarquay C, Réthoré G, Benadjaoud MA, Siñeriz F, Pattapa G, Guicheux J, Weiss P, Barritault D, Mathieu N. Heparan Sulfate Mimetics: A New Way to Optimize Therapeutic Effects of Hydrogel-Embedded Mesenchymal Stromal Cells in Colonic Radiation-Induced Damage. Sci Rep 2019; 9:164. [PMID: 30655576 PMCID: PMC6336771 DOI: 10.1038/s41598-018-36631-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/25/2018] [Indexed: 12/18/2022] Open
Abstract
Clinical expression of gastrointestinal radiation toxicity on non-cancerous tissue could be very life threatening and clinicians must deal increasingly with the management of late side effects of radiotherapy. Cell therapy, in particular mesenchymal stromal cell (MSC) therapy, has shown promising results in numerous preclinical animal studies and thus has emerged as a new hope for patient refractory to current treatments. However, many stem cell clinical trials do not confer any beneficial effect suggesting a real need to accelerate research towards the successful clinical application of stem cell therapy. In this study, we propose a new concept to improve the procedure of MSC-based treatment for greater efficacy and clinical translatability. We demonstrated that heparan sulfate mimetic (HS-m) injections that restore the extracellular matrix network and enhance the biological activity of growth factors, associated with local injection of MSC protected in a hydrogel, that increase cell engraftment and cell survival, improve the therapeutic benefit of MSC treatment in two animal models relevant of the human pathology. For the first time, a decrease of the injury score in the ulcerated area was observed with this combined treatment. We also demonstrated that the combined treatment favored the epithelial regenerative process. In this study, we identified a new way, clinically applicable, to optimize stem-cell therapy and could be proposed to patients suffering from severe colonic defect after radiotherapy.
Collapse
Affiliation(s)
- Lara Moussa
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, PSE-SANTE, SERAMED, LRMed, 31 avenue de la division Leclerc, 92262, Fontenay-aux-Roses, France.,INSERM, Institut National de la Santé et de la Recherche Médicale, U1229, Regenerative Medicine and Skeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France.,Université de Nantes, Regenerative Medicine and Squeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France
| | - Christelle Demarquay
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, PSE-SANTE, SERAMED, LRMed, 31 avenue de la division Leclerc, 92262, Fontenay-aux-Roses, France
| | - Gildas Réthoré
- INSERM, Institut National de la Santé et de la Recherche Médicale, U1229, Regenerative Medicine and Skeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France.,Université de Nantes, Regenerative Medicine and Squeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France.,Centre Hospitalier Universitaire de Nantes, Pôle Hospitalo-Universitaire 4 (OTONN), 1 Place Alexis Ricordeau, 44042, Nantes, France
| | - Mohamed Amine Benadjaoud
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, PSE-SANTE, SERAMED, LRMed, 31 avenue de la division Leclerc, 92262, Fontenay-aux-Roses, France
| | - Fernando Siñeriz
- Société OTR3 (Organes, Tissus, Régénération, Réparation, Remplacement), 4 Rue Française, 75001, Paris, France
| | - Girish Pattapa
- INSERM, Institut National de la Santé et de la Recherche Médicale, U1229, Regenerative Medicine and Skeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France.,Université de Nantes, Regenerative Medicine and Squeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France
| | - Jérôme Guicheux
- INSERM, Institut National de la Santé et de la Recherche Médicale, U1229, Regenerative Medicine and Skeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France.,Université de Nantes, Regenerative Medicine and Squeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France.,Centre Hospitalier Universitaire de Nantes, Pôle Hospitalo-Universitaire 4 (OTONN), 1 Place Alexis Ricordeau, 44042, Nantes, France
| | - Pierre Weiss
- INSERM, Institut National de la Santé et de la Recherche Médicale, U1229, Regenerative Medicine and Skeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France.,Université de Nantes, Regenerative Medicine and Squeleton (RMeS), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042, Nantes, France.,Centre Hospitalier Universitaire de Nantes, Pôle Hospitalo-Universitaire 4 (OTONN), 1 Place Alexis Ricordeau, 44042, Nantes, France
| | - Denis Barritault
- Société OTR3 (Organes, Tissus, Régénération, Réparation, Remplacement), 4 Rue Française, 75001, Paris, France.,Université Paris-Est Créteil, Laboratoire de recherche sur la Croissance Cellulaire, Réparation, et Régénération Tissulaire, Faculté des Sciences, Université Paris-Est Créteil, 61 Ave du Gal de Gaulle, 94000, Créteil, France
| | - Noëlle Mathieu
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, PSE-SANTE, SERAMED, LRMed, 31 avenue de la division Leclerc, 92262, Fontenay-aux-Roses, France.
| |
Collapse
|
27
|
Khan K, Gasbarrino K, Mahmoud I, Makhoul G, Yu B, Dufresne L, Daskalopoulou SS, Schwertani A, Cecere R. Bioactive scaffolds in stem-cell-based therapies for cardiac repair: protocol for a meta-analysis of randomized controlled preclinical trials in animal myocardial infarction models. Syst Rev 2018; 7:225. [PMID: 30518435 PMCID: PMC6280361 DOI: 10.1186/s13643-018-0845-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/17/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Acute myocardial infarction (MI) remains one of the leading causes of death worldwide with no curative therapy available. Stem cell therapies have been gaining interest as a means to repair the cardiac tissue after MI and prevent the onset of heart failure. Many in vivo reports suggest that the use of stem cells is promising, yet clinical trials suggest that the cells fail to integrate into the native tissue, resulting in limited improvements in cardiac function and repair. To battle this limitation, the combination of using stem cells embedded in a bioactive scaffold that promotes cell retention is growing in interest. Yet, a systematic review of the literature on the use of stem cells embedded in bioactive scaffolds for cardiac repair has not yet been performed. In this protocol, we outline a systematic review and meta-analysis of preclinical trials in animal MI models that utilize stem cell-embedded scaffolds for cardiac repair and compare their effects to stem cell-treated animals without the use of a scaffold. METHODS/DESIGN We will search the following electronic databases: Cochrane Library, MEDLINE, Embase, PubMed, Scopus and Web of Science, and gray literature: Canadian Agency for Drugs and Technologies in Health and Google Scholar. We will only include randomly controlled preclinical trials that have directly investigated the effects of stem cells embedded in a scaffold for cardiac repair in an animal MI model. Two investigators will independently review each article included in the final analysis. The primary endpoint that will be investigated is left ventricular ejection fraction. Secondary endpoints will include infarct size, end systolic volume, end diastolic volume, fractional shortening and left ventricular wall thickness. Pooled analyses will be conducted using the DerSimonian-Laird random effects and Mantel-Haenszel fixed-effect models. Between-studies heterogeneity will be quantified and determined using the Tau2 and I2 statistics. Publication bias will be assessed using visual inspection of funnel plots and complemented by Begg's and Egger's statistical tests. Possible sources of heterogeneity will be assessed using subgroup-meta analysis and meta-regression. DISCUSSION To date, the use of scaffolds in myocardial repair has not yet been systematically reviewed. The results of this meta-analysis will aid in determining the efficacy of stem cell-embedded scaffolds for cardiac repair and help bring this therapy to the clinic.
Collapse
Affiliation(s)
- Kashif Khan
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec Canada
| | - Karina Gasbarrino
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, McGill University Health Centre, Montreal, Quebec Canada
| | - Ibtisam Mahmoud
- McConnell Resource Centre, McGill University Health Centre, Montreal, Quebec Canada
| | - Georges Makhoul
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec Canada
| | - Bin Yu
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec Canada
| | - Line Dufresne
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec Canada
| | - Stella S. Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, McGill University Health Centre, Montreal, Quebec Canada
| | - Adel Schwertani
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec Canada
| | - Renzo Cecere
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec Canada
- Glen Campus-The Royal Victoria Hospital, 1001 Decarie Blvd, Block C, C07.1284, Montreal, Quebec H4A 3J1 Canada
| |
Collapse
|
28
|
In situ photochemical crosslinking of hydrogel membrane for Guided Tissue Regeneration. Dent Mater 2018; 34:1769-1782. [PMID: 30336953 DOI: 10.1016/j.dental.2018.09.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Periodontitis is an inflammatory disease that destroys the tooth-supporting attachment apparatus. Guided tissue regeneration (GTR) is a technique based on a barrier membrane designed to prevent wound space colonization by gingival cells. This study examined a new formulation composed of two polymers that could be photochemically cross-linked in situ into an interpenetrated polymer network (IPN) forming a hydrogel membrane. METHODS We synthetized and characterized silanized hydroxypropyl methylcellulose (Si-HPMC) for its cell barrier properties and methacrylated carboxymethyl chitosan (MA-CMCS) for its degradable backbone to use in IPN. Hydrogel membranes were cross-linked using riboflavin photoinitiator and a dentistry visible light lamp. The biomaterial's physicochemical and mechanical properties were determined. Hydrogel membrane degradation was evaluated in lysozyme. Cytocompatibility was estimated by neutral red uptake. The cell barrier property was studied culturing human primary gingival fibroblasts or human gingival explants on membrane and analyzed with confocal microscopy and histological staining. RESULTS The IPN hydrogel membrane was obtained after 120s of irradiation. The IPN showed a synergistic increase in Young moduli compared with the single networks. The CMCS addition in IPN allows a progressive weight loss compared to each polymer network. Cytocompatibility was confirmed by neutral red assay. Human cell invasion was prevented by hydrogel membranes and histological sections revealed that the biomaterial exhibited a barrier effect in contact with soft gingival tissue. SIGNIFICANCE We demonstrated the ability of an innovative polymer formulation to form in situ, using a dentist's lamp, an IPN hydrogel membrane, which could be an easy-to-use biomaterial for GTR therapy.
Collapse
|
29
|
The use of hydrogels for cell-based treatment of chronic kidney disease. Clin Sci (Lond) 2018; 132:1977-1994. [PMID: 30220651 DOI: 10.1042/cs20180434] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/01/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is a major and growing public health concern with increasing incidence and prevalence worldwide. The therapeutic potential of stem cell therapy, including mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) holds great promise for treatment of CKD. However, there are significant bottlenecks in the clinical translation due to the reduced number of transplanted cells and the duration of their presence at the site of tissue damage. Bioengineered hydrogels may provide a route of cell delivery to enhance treatment efficacy and optimise the targeting effectiveness while minimising any loss of cell function. In this review, we highlight the advances in stem cell therapy targeting kidney disease and discuss the emerging role of hydrogel delivery systems to fully realise the potential of adult stem cells as a regenerative therapy for CKD in humans. MSCs and EPCs mediate kidney repair through distinct paracrine effects. As a delivery system, hydrogels can prolong these paracrine effects by improving retention at the site of injury and protecting the transplanted cells from the harsh inflammatory microenvironment. We also discuss the features of a hydrogel, which may be tuned to optimise the therapeutic potential of encapsulated stem cells, including cell-adhesive epitopes, material stiffness, nanotopography, modes of gelation and degradation and the inclusion of bioactive molecules. This review concludes with a discussion of the challenges to be met for the widespread clinical use of hydrogel delivery system of stem cell therapy for CKD.
Collapse
|
30
|
Sustained release of targeted cardiac therapy with a replenishable implanted epicardial reservoir. Nat Biomed Eng 2018; 2:416-428. [DOI: 10.1038/s41551-018-0247-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 05/09/2018] [Indexed: 12/12/2022]
|
31
|
Ciuffreda MC, Malpasso G, Chokoza C, Bezuidenhout D, Goetsch KP, Mura M, Pisano F, Davies NH, Gnecchi M. Synthetic extracellular matrix mimic hydrogel improves efficacy of mesenchymal stromal cell therapy for ischemic cardiomyopathy. Acta Biomater 2018; 70:71-83. [PMID: 29341932 DOI: 10.1016/j.actbio.2018.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 12/28/2017] [Accepted: 01/08/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND Mesenchymal stromal cells (MSC) repair infarcted hearts mainly through paracrine mechanisms. Low cell engraftment limits the release of soluble paracrine factors (SF) over time and, consequently, MSC efficacy. We tested whether a synthetic extracellular matrix mimic, a hydrogel containing heparin (H-HG), could ameliorate MSC engraftment and binding/release of SF, thus improving MSC therapy efficacy. METHODS AND RESULTS In vitro, rat bone-marrow MSC (rBM-MSC) were seeded and grown into H-HG. Under normoxia, the hydrogel did not affect cell survival (rBM-MSC survival >90% at each time point tested); vice versa, under hypoxia the biomaterial resulted to be protective for the cells (p < .001 vs rBM-MSC alone). H-HG or control PEG hydrogels (HG) were incubated with VEGF or bFGF for binding/release quantification. Data showed significantly higher amount of VEGF and bFGF bound by H-HG compared with HG (p < .05) and a constant release over time. In vivo, myocardial infarction (MI) was induced in female Sprague Dawley rats by permanent coronary ligation. One week later, saline, rBM-MSC, H-HG or rBM-MSC/H-HG were injected in the infarct zone. The co-injection of rBM-MSC/H-HG into infarcted hearts significantly increased cardiac function. Importantly, we observed a significant gain in MSC engraftment, reduction of ventricular remodeling and stimulation of neo-vasculogenesis. We also documented higher amounts of several pro-angiogenic factors in hearts treated with rBM-MSC/H-HG. CONCLUSIONS Our data show that H-HG increases MSC engraftment, efficiently fine tunes the paracrine MSC actions and improves cardiac function in infarcted rat hearts. STATEMENT OF SIGNIFICANCE Transplantation of MSC is a promising treatment for ischemic heart disease, but low cell engraftment has so far limited its efficacy. The enzymatically degradable H-HG that we developed is able to increase MSC retention/engraftment and, at the same time, to fine-tune the paracrine effects mediated by the cells. Most importantly, the co-transplantation of MSC and H-HG in a rat model of ischemic cardiomyopathy improved heart function through a significant reduction in ventricular remodeling/scarring and amelioration in neo-vasculogenesis/endogenous cardiac regeneration. These beneficial effects are comparable to those obtained by others using a much greater number of cells, strengthening the efficacy of the biomaterial used in increasing the therapeutic effects of MSC. Given its efficacy and safety, documented by the absence of immunoreaction, our strategy appears readily translatable to clinical scenarios.
Collapse
Affiliation(s)
- Maria Chiara Ciuffreda
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy
| | - Giuseppe Malpasso
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy
| | - Cindy Chokoza
- Cardiovascular Research Unit, Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Department of Health Sciences, Cape Town, South Africa
| | - Deon Bezuidenhout
- Cardiovascular Research Unit, Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Department of Health Sciences, Cape Town, South Africa
| | - Kyle P Goetsch
- Cardiovascular Research Unit, Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Department of Health Sciences, Cape Town, South Africa
| | - Manuela Mura
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy
| | - Federica Pisano
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy
| | - Neil H Davies
- Cardiovascular Research Unit, Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Department of Health Sciences, Cape Town, South Africa
| | - Massimiliano Gnecchi
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy; Department of Medicine, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
32
|
Figueiredo L, Pace R, D'Arros C, Réthoré G, Guicheux J, Le Visage C, Weiss P. Assessing glucose and oxygen diffusion in hydrogels for the rational design of 3D stem cell scaffolds in regenerative medicine. J Tissue Eng Regen Med 2018; 12:1238-1246. [DOI: 10.1002/term.2656] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 01/17/2018] [Accepted: 02/17/2018] [Indexed: 12/14/2022]
Affiliation(s)
- L. Figueiredo
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton; Université de Nantes, ONIRIS; Nantes France
- UFR Odontologie; Université de Nantes; Nantes France
| | - R. Pace
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton; Université de Nantes, ONIRIS; Nantes France
- UFR Odontologie; Université de Nantes; Nantes France
| | - C. D'Arros
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton; Université de Nantes, ONIRIS; Nantes France
- UFR Odontologie; Université de Nantes; Nantes France
| | - G. Réthoré
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton; Université de Nantes, ONIRIS; Nantes France
- UFR Odontologie; Université de Nantes; Nantes France
| | - J. Guicheux
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton; Université de Nantes, ONIRIS; Nantes France
- UFR Odontologie; Université de Nantes; Nantes France
- CHU Nantes, PHU 4 OTONN; Nantes France
| | - C. Le Visage
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton; Université de Nantes, ONIRIS; Nantes France
- UFR Odontologie; Université de Nantes; Nantes France
| | - P. Weiss
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton; Université de Nantes, ONIRIS; Nantes France
- UFR Odontologie; Université de Nantes; Nantes France
- CHU Nantes, PHU 4 OTONN; Nantes France
| |
Collapse
|
33
|
Retention and Functional Effect of Adipose-Derived Stromal Cells Administered in Alginate Hydrogel in a Rat Model of Acute Myocardial Infarction. Stem Cells Int 2018; 2018:7821461. [PMID: 29765421 PMCID: PMC5892231 DOI: 10.1155/2018/7821461] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/05/2018] [Accepted: 01/18/2018] [Indexed: 01/12/2023] Open
Abstract
Background Cell therapy for heart disease has been proven safe and efficacious, despite poor cell retention in the injected area. Improving cell retention is hypothesized to increase the treatment effect. In the present study, human adipose-derived stromal cells (ASCs) were delivered in an in situ forming alginate hydrogel following acute myocardial infarction (AMI) in rats. Methods ASCs were transduced with luciferase and tested for ASC phenotype. AMI was inducted in nude rats, with subsequent injection of saline (controls), 1 × 106 ASCs in saline or 1 × 106 ASCs in 1% (w/v) alginate hydrogel. ASCs were tracked by bioluminescence and functional measurements were assessed by magnetic resonance imaging (MRI) and 82rubidium positron emission tomography (PET). Results ASCs in both saline and alginate hydrogel significantly increased the ejection fraction (7.2% and 7.8% at 14 days and 7.2% and 8.0% at 28 days, resp.). After 28 days, there was a tendency for decreased infarct area and increased perfusion, compared to controls. No significant differences were observed between ASCs in saline or alginate hydrogel, in terms of retention and functional salvage. Conclusion ASCs improved the myocardial function after AMI, but administration in the alginate hydrogel did not further improve retention of the cells or myocardial function.
Collapse
|
34
|
Moussa L, Usunier B, Demarquay C, Benderitter M, Tamarat R, Sémont A, Mathieu N. Bowel Radiation Injury: Complexity of the Pathophysiology and Promises of Cell and Tissue Engineering. Cell Transplant 2018; 25:1723-1746. [PMID: 27197023 DOI: 10.3727/096368916x691664] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ionizing radiation is effective to treat malignant pelvic cancers, but the toxicity to surrounding healthy tissue remains a substantial limitation. Early and late side effects not only limit the escalation of the radiation dose to the tumor but may also be life-threatening in some patients. Numerous preclinical studies determined specific mechanisms induced after irradiation in different compartments of the intestine. This review outlines the complexity of the pathogenesis, highlighting the roles of the epithelial barrier in the vascular network, and the inflammatory microenvironment, which together lead to chronic fibrosis. Despite the large number of pharmacological molecules available, the studies presented in this review provide encouraging proof of concept regarding the use of mesenchymal stromal cell (MSC) therapy to treat radiation-induced intestinal damage. The therapeutic efficacy of MSCs has been demonstrated in animal models and in patients, but an enormous number of cells and multiple injections are needed due to their poor engraftment capacity. Moreover, it has been observed that although MSCs have pleiotropic effects, some intestinal compartments are less restored after a high dose of irradiation. Future research should seek to optimize the efficacy of the injected cells, particularly with regard to extending their life span in the irradiated tissue. Moreover, improving the host microenvironment, combining MSCs with other specific regenerative cells, or introducing new tissue engineering strategies could be tested as methods to treat the severe side effects of pelvic radiotherapy.
Collapse
Affiliation(s)
- Lara Moussa
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Benoît Usunier
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Christelle Demarquay
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Marc Benderitter
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Radia Tamarat
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Alexandra Sémont
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Noëlle Mathieu
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| |
Collapse
|
35
|
Montheil T, Echalier C, Martinez J, Subra G, Mehdi A. Inorganic polymerization: an attractive route to biocompatible hybrid hydrogels. J Mater Chem B 2018; 6:3434-3448. [DOI: 10.1039/c8tb00456k] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The sol–gel process is one of the main techniques leading to hybrid hydrogels that can be used in a wide scope of applications, especially in the biomedical field.
Collapse
Affiliation(s)
- Titouan Montheil
- Institut des Biomolécules Max Mousseron
- Université de Montpellier
- CNRS
- ENSCM
- Montpellier
| | - Cécile Echalier
- Institut des Biomolécules Max Mousseron
- Université de Montpellier
- CNRS
- ENSCM
- Montpellier
| | - Jean Martinez
- Institut des Biomolécules Max Mousseron
- Université de Montpellier
- CNRS
- ENSCM
- Montpellier
| | - Gilles Subra
- Institut des Biomolécules Max Mousseron
- Université de Montpellier
- CNRS
- ENSCM
- Montpellier
| | - Ahmad Mehdi
- Institut Charles Gerhardt Université de Montpellier
- CNRS
- ENSCM
- Montpellier
- France
| |
Collapse
|
36
|
Cellular Therapeutics for Heart Failure: Focus on Mesenchymal Stem Cells. Stem Cells Int 2017; 2017:9640108. [PMID: 29391871 PMCID: PMC5748110 DOI: 10.1155/2017/9640108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/31/2017] [Accepted: 08/14/2017] [Indexed: 12/28/2022] Open
Abstract
Resulting from a various etiologies, the most notable remains ischemia; heart failure (HF) manifests as the common end pathway of many cardiovascular processes and remains among the top causes for hospitalization and a major cause of morbidity and mortality worldwide. Current pharmacologic treatment for HF utilizes pharmacologic agents to control symptoms and slow further deterioration; however, on a cellular level, in a patient with progressive disease, fibrosis and cardiac remodeling can continue leading to end-stage heart failure. Cellular therapeutics have risen as the new hope for an improvement in the treatment of HF. Mesenchymal stem cells (MSCs) have gained popularity given their propensity of promoting endogenous cellular repair of a myriad of disease processes via paracrine signaling through expression of various cytokines, chemokines, and adhesion molecules resulting in activation of signal transduction pathways. While the exact mechanism remains to be completely elucidated, this remains the primary mechanism identified to date. Recently, MSCs have been incorporated as the central focus in clinical trials investigating the role how MSCs can play in the treatment of HF. In this review, we focus on the characteristics of MSCs that give them a distinct edge as cellular therapeutics and present results of clinical trials investigating MSCs in the setting of ischemic HF.
Collapse
|
37
|
Henry N, Clouet J, Le Bideau J, Le Visage C, Guicheux J. Innovative strategies for intervertebral disc regenerative medicine: From cell therapies to multiscale delivery systems. Biotechnol Adv 2017; 36:281-294. [PMID: 29199133 DOI: 10.1016/j.biotechadv.2017.11.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 12/14/2022]
Abstract
As our understanding of the physiopathology of intervertebral disc (IVD) degeneration has improved, novel therapeutic strategies have emerged, based on the local injection of cells, bioactive molecules, and nucleic acids. However, with regard to the harsh environment constituted by degenerated IVDs, protecting biologics from in situ degradation while allowing their long-term delivery is a major challenge. Yet, the design of the optimal approach for IVD regeneration is still under debate and only a few papers provide a critical assessment of IVD-specific carriers for local and sustained delivery of biologics. In this review, we highlight the IVD-relevant polymers as well as their design as macro-, micro-, and nano-sized particles to promote endogenous repair. Finally, we illustrate how multiscale systems, combining in situ-forming hydrogels with ready-to-use particles, might drive IVD regenerative medicine strategies toward innovation.
Collapse
Affiliation(s)
- Nina Henry
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France; Institut des Matériaux Jean Rouxel (IMN), Université de Nantes, CNRS, 2 rue de la Houssinière, BP 32229, 44322 Nantes, Cedex 3, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France
| | - Johann Clouet
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France; CHU Nantes, Pharmacie Centrale, PHU 11, Nantes, France; Université de Nantes, UFR Sciences Biologiques et Pharmaceutiques, Nantes, France
| | - Jean Le Bideau
- Institut des Matériaux Jean Rouxel (IMN), Université de Nantes, CNRS, 2 rue de la Houssinière, BP 32229, 44322 Nantes, Cedex 3, France
| | - Catherine Le Visage
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France.
| | - Jérôme Guicheux
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France; CHU Nantes, PHU 4 OTONN, Nantes, France.
| |
Collapse
|
38
|
Henry N, Clouet J, Fragale A, Griveau L, Chédeville C, Véziers J, Weiss P, Le Bideau J, Guicheux J, Le Visage C. Pullulan microbeads/Si-HPMC hydrogel injectable system for the sustained delivery of GDF-5 and TGF-β1: new insight into intervertebral disc regenerative medicine. Drug Deliv 2017; 24:999-1010. [PMID: 28645219 PMCID: PMC8241148 DOI: 10.1080/10717544.2017.1340362] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/31/2017] [Accepted: 06/06/2017] [Indexed: 12/16/2022] Open
Abstract
Discogenic low back pain is considered a major health concern and no etiological treatments are today available to tackle this disease. To clinically address this issue at early stages, there is a rising interest in the stimulation of local cells by in situ injection of growth factors targeting intervertebral disc (IVD) degenerative process. Despite encouraging safety and tolerability results in clinic, growth factors efficacy may be further improved. To this end, the use of a delivery system allowing a sustained release, while protecting growth factors from degradation appears of particular interest. We propose herein the design of a new injectable biphasic system, based on the association of pullulan microbeads (PMBs) into a cellulose-based hydrogel (Si-HPMC), for the TGF-β1 and GDF-5 growth factors sustained delivery. We present for the first time the design and mechanical characterization of both the PMBs and the called biphasic system (PMBs/Si-HPMC). Their loading and release capacities were also studied and we were able to demonstrate a sustained release of both growth factors, for up to 28 days. Noteworthy, the growth factors biological activity on human cells was maintained. Altogether, these data suggest that this PMBs/Si-HPMC biphasic system may be a promising candidate for the development of an innovative bioactive delivery system for IVD regenerative medicine.
Collapse
Affiliation(s)
- Nina Henry
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- Institut des Matériaux Jean Rouxel (IMN), Université de Nantes, CNRS, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
| | - Johann Clouet
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
- CHU Nantes, PHU 11 Pharmacie, Pharmacie Centrale, Nantes, France
- UFR Sciences Biologiques et Pharmaceutiques, Université de Nantes, Nantes, France
| | - Audrey Fragale
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
| | - Louise Griveau
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
| | - Claire Chédeville
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
| | - Joëlle Véziers
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
- SC3M platform, UMS INSERM 016/CNRS 3556, SFR François Bonamy, Nantes, France
- CHU Nantes, PHU 4 OTONN, Nantes, France
| | - Pierre Weiss
- UFR Odontologie, Université de Nantes, Nantes, France
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team REGOS “Regenerative Medicine of Bone Tissues”, Nantes, France
| | - Jean Le Bideau
- Institut des Matériaux Jean Rouxel (IMN), Université de Nantes, CNRS, Nantes, France
| | - Jérôme Guicheux
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
- CHU Nantes, PHU 4 OTONN, Nantes, France
| | - Catherine Le Visage
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
| |
Collapse
|
39
|
Wu YC, Wan ZH, Rong YH, Zhu B, Jiang QY, Liu HL, Li DZ, Zhao J, Xing HQ, Su HB, Hu JH, Xin SJ, Zang H, You SL. Bioactivity of CD34+ cells in patients with acute-on-chronic liver failure. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:10781-10791. [PMID: 31966421 PMCID: PMC6965859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 02/25/2017] [Indexed: 06/10/2023]
Abstract
Liver failure is a life-threatened serious disease with many complications and high mortality rate. Stem cells have been applied to replacement therapy, gene therapy and tissue engineering for its capacity of self-renewal and multi-lineage differentiation. To investigate the bioactivity of the peripheral blood hematopoietic stem cells (PBHSC) in patients with acute-on-chronic liver failure, we isolated CD34+ cells from peripheral blood of patients with acute-on-chronic liver failure and healthy controls. After cultured it in serum-free medium (SFEM), we studied the bioactivity of CD34+ cells by observing the morphology, recording growth curve, detecting cell cycle and cell apoptosis. CD34+ cells and culture solution were collected at the time points of 3, 5, 7, 10, 12 and 14 days, and the levels of hepatocyte growth factor (HGF), matrix metalloproteinase-9 (MMP-9), tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in culture solution were detected by ELISA. Also, the expressions of pyruvate kinase muscle isoenzyme 2 (PKM2), integrin-β1 and liver-type pyruvate kinase (LPK) were detected by RT-PCR and immunofluorescence. Our results showed the bioactivity of CD34+ cells from patients with acute-on-chronic liver failure was identified to be similar with that from healthy controls. HGF, MMP-9, TNF-α and IL-6 were found in cell culture medium. RT-PCR and immunofluorescence results indicated that PKM2, Integrin-β1 expressed on CD34+ cells from patients with acute-on-chronic liver failure. In conclusion, bioactivity of CD34+ cells of patients with acute-on-chronic liver failure was demonstrated to be normal, which could secrete HGF, MMP-9, TNF-α and IL-6, promote the growth of hepatocytes, and differentiate along a direction to hepatocyte lineage.
Collapse
Affiliation(s)
- Yi-Chen Wu
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| | - Zhi-Hong Wan
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| | - Yi-Hui Rong
- Treatment and Research Center for Liver Cancer, 302 Hospital of PLABeijing, China
| | - Bing Zhu
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| | - Qi-Yu Jiang
- Research Center for Clinical Medicine, 302 Hospital of PLABeijing, China
| | - Hong-Ling Liu
- Liver Transplantation Research Center, 302 Hospital of PLABeijing, China
| | - Dong-Ze Li
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| | - Jun Zhao
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| | - Han-Qian Xing
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| | - Hai-Bin Su
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| | - Jin-Hua Hu
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| | - Shao-Jie Xin
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| | - Hong Zang
- Liver Transplantation Research Center, 302 Hospital of PLABeijing, China
| | - Shao-Li You
- Liver Failure Treatment and Research Center, 302 Hospital of PLABeijing, China
| |
Collapse
|
40
|
Flégeau K, Pace R, Gautier H, Rethore G, Guicheux J, Le Visage C, Weiss P. Toward the development of biomimetic injectable and macroporous biohydrogels for regenerative medicine. Adv Colloid Interface Sci 2017; 247:589-609. [PMID: 28754381 DOI: 10.1016/j.cis.2017.07.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/13/2017] [Accepted: 07/13/2017] [Indexed: 01/21/2023]
Abstract
Repairing or replacing damaged human tissues has been the ambitious goal of regenerative medicine for over 25years. One promising approach is the use of hydrated three-dimensional scaffolds, known as hydrogels, which have had good results repairing tissues in pre-clinical trials. Benefiting from breakthrough advances in the field of biology, and more particularly regarding cell/matrix interactions, these hydrogels are now designed to recapitulate some of the fundamental cues of native environments to drive the local tissue regeneration. We highlight the key parameters that are required for the development of smart and biomimetic hydrogels. We also review the wide variety of polymers, crosslinking methods, and manufacturing processes that have been developed over the years. Of particular interest is the emergence of supramolecular chemistries, allowing for the development of highly functional and reversible biohydrogels. Moreover, advances in computer assisted design and three-dimensional printing have revolutionized the production of macroporous hydrogels and allowed for more complex designs than ever before with the opportunity to develop fully reconstituted organs. Today, the field of biohydrogels for regenerative medicine is a prolific area of research with applications for most bodily tissues. On top of these applications, injectable hydrogels and macroporous hydrogels (foams) were found to be the most successful. While commonly associated with cells or biologics as drug delivery systems to increase therapeutic outcomes, they are steadily being used in the emerging fields of organs-on-chip and hydrogel-assisted cell therapy. To highlight these advances, we review some of the recent developments that have been achieved for the regeneration of tissues, focusing on the articular cartilage, bone, cardiac, and neural tissues. These biohydrogels are associated with improved cartilage and bone defects regeneration, reduced left ventricular dilation upon myocardial infarction and display promising results repairing neural lesions. Combining the benefits from each of these areas reviewed above, we envision that an injectable biohydrogel foam loaded with either stem cells or their secretome is the most promising hydrogel solution to trigger tissue regeneration. A paradigm shift is occurring where the combined efforts of fundamental and applied sciences head toward the development of hydrogels restoring tissue functions, serving as drug screening platforms or recreating complex organs.
Collapse
|
41
|
Mayourian J, Cashman TJ, Ceholski DK, Johnson BV, Sachs D, Kaji DA, Sahoo S, Hare JM, Hajjar RJ, Sobie EA, Costa KD. Experimental and Computational Insight Into Human Mesenchymal Stem Cell Paracrine Signaling and Heterocellular Coupling Effects on Cardiac Contractility and Arrhythmogenicity. Circ Res 2017. [PMID: 28642329 DOI: 10.1161/circresaha.117.310796] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Myocardial delivery of human mesenchymal stem cells (hMSCs) is an emerging therapy for treating the failing heart. However, the relative effects of hMSC-mediated heterocellular coupling (HC) and paracrine signaling (PS) on human cardiac contractility and arrhythmogenicity remain unresolved. OBJECTIVE The objective is to better understand hMSC PS and HC effects on human cardiac contractility and arrhythmogenicity by integrating experimental and computational approaches. METHODS AND RESULTS Extending our previous hMSC-cardiomyocyte HC computational model, we incorporated experimentally calibrated hMSC PS effects on cardiomyocyte L-type calcium channel/sarcoendoplasmic reticulum calcium-ATPase activity and cardiac tissue fibrosis. Excitation-contraction simulations of hMSC PS-only and combined HC+PS effects on human cardiomyocytes were representative of human engineered cardiac tissue (hECT) contractile function measurements under matched experimental treatments. Model simulations and hECTs both demonstrated that hMSC-mediated effects were most pronounced under PS-only conditions, where developed force increased ≈4-fold compared with non-hMSC-supplemented controls during physiological 1-Hz pacing. Simulations predicted contractility of isolated healthy and ischemic adult human cardiomyocytes would be minimally sensitive to hMSC HC, driven primarily by PS. Dominance of hMSC PS was also revealed in simulations of fibrotic cardiac tissue, where hMSC PS protected from potential proarrhythmic effects of HC at various levels of engraftment. Finally, to study the nature of the hMSC paracrine effects on contractility, proteomic analysis of hECT/hMSC conditioned media predicted activation of PI3K/Akt signaling, a recognized target of both soluble and exosomal fractions of the hMSC secretome. Treating hECTs with exosome-enriched, but not exosome-depleted, fractions of the hMSC secretome recapitulated the effects observed with hMSC conditioned media on hECT-developed force and expression of calcium-handling genes (eg, SERCA2a, L-type calcium channel). CONCLUSIONS Collectively, this integrated experimental and computational study helps unravel relative hMSC PS and HC effects on human cardiac contractility and arrhythmogenicity, and provides novel insight into the role of exosomes in hMSC paracrine-mediated effects on contractility.
Collapse
Affiliation(s)
- Joshua Mayourian
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Timothy J Cashman
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Delaine K Ceholski
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Bryce V Johnson
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - David Sachs
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Deepak A Kaji
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Susmita Sahoo
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Joshua M Hare
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Roger J Hajjar
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Eric A Sobie
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Kevin D Costa
- From the Cardiovascular Research Center (J.M., T.J.C., D.K.C., D.S., S.S., R.J.H., K.D.C.), Department of Developmental and Regenerative Biology (D.A.K.), and Department of Pharmacology and Systems Therapeutics (E.A.S.), Icahn School of Medicine at Mount Sinai, New York; Department of Medicine, University of Washington Seattle (B.V.J.); and The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H.).
| |
Collapse
|
42
|
Wobma HM, Liu D, Vunjak-Novakovic G. Paracrine Effects of Mesenchymal Stromal Cells Cultured in Three-Dimensional Settings on Tissue Repair. ACS Biomater Sci Eng 2017; 4:1162-1175. [PMID: 33418654 DOI: 10.1021/acsbiomaterials.7b00005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a promising cell source for promoting tissue repair, due to their ability to release growth, angiogenic, and immunomodulatory factors. However, when injected as a suspension, these cells suffer from poor survival and localization, and suboptimal release of paracrine factors. While there have been attempts to overcome these limitations by modifying MSCs themselves, a more versatile solution is to grow them in three dimensions, as aggregates or embedded into biomaterials. Here we review the mechanisms by which 3D culture can influence the regenerative capacity of undifferentiated MSCs, focusing on recent examples from the literature. We further discuss how knowledge of these mechanisms can lead to strategic design of MSC therapies that overcome some of the challenges to their effective translation.
Collapse
|
43
|
Hydrogel based approaches for cardiac tissue engineering. Int J Pharm 2017; 523:454-475. [DOI: 10.1016/j.ijpharm.2016.10.061] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/24/2016] [Accepted: 10/26/2016] [Indexed: 01/04/2023]
|
44
|
Passipieri JA, Baker HB, Siriwardane M, Ellenburg MD, Vadhavkar M, Saul JM, Tomblyn S, Burnett L, Christ GJ. Keratin Hydrogel Enhances In Vivo Skeletal Muscle Function in a Rat Model of Volumetric Muscle Loss. Tissue Eng Part A 2017; 23:556-571. [PMID: 28169594 DOI: 10.1089/ten.tea.2016.0458] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Volumetric muscle loss (VML) injuries exceed the considerable intrinsic regenerative capacity of skeletal muscle, resulting in permanent functional and cosmetic deficits. VML and VML-like injuries occur in military and civilian populations, due to trauma and surgery as well as due to a host of congenital and acquired diseases/syndromes. Current therapeutic options are limited, and new approaches are needed for a more complete functional regeneration of muscle. A potential solution is human hair-derived keratin (KN) biomaterials that may have significant potential for regenerative therapy. The goal of these studies was to evaluate the utility of keratin hydrogel formulations as a cell and/or growth factor delivery vehicle for functional muscle regeneration in a surgically created VML injury in the rat tibialis anterior (TA) muscle. VML injuries were treated with KN hydrogels in the absence and presence of skeletal muscle progenitor cells (MPCs), and/or insulin-like growth factor 1 (IGF-1), and/or basic fibroblast growth factor (bFGF). Controls included VML injuries with no repair (NR), and implantation of bladder acellular matrix (BAM, without cells). Initial studies conducted 8 weeks post-VML injury indicated that application of keratin hydrogels with growth factors (KN, KN+IGF-1, KN+bFGF, and KN+IGF-1+bFGF, n = 8 each) enabled a significantly greater functional recovery than NR (n = 7), BAM (n = 8), or the addition of MPCs to the keratin hydrogel (KN+MPC, KN+MPC+IGF-1, KN+MPC+bFGF, and KN+MPC+IGF-1+bFGF, n = 8 each) (p < 0.05). A second series of studies examined functional recovery for as many as 12 weeks post-VML injury after application of keratin hydrogels in the absence of cells. A significant time-dependent increase in functional recovery of the KN, KN+bFGF, and KN+IGF+bFGF groups was observed, relative to NR and BAM implantation, achieving as much as 90% of the maximum possible functional recovery. Histological findings from harvested tissue at 12 weeks post-VML injury documented significant increases in neo-muscle tissue formation in all keratin treatment groups as well as diminished fibrosis, in comparison to both BAM and NR. In conclusion, keratin hydrogel implantation promoted statistically significant and physiologically relevant improvements in functional outcomes post-VML injury to the rodent TA muscle.
Collapse
Affiliation(s)
- J A Passipieri
- 1 Biomedical Engineering Department, University of Virginia , Charlottesville, Virginia.,2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina
| | - H B Baker
- 2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina.,3 Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| | - Mevan Siriwardane
- 2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina
| | | | - Manasi Vadhavkar
- 2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina
| | - Justin M Saul
- 5 Department of Chemical, Paper and Biomedical Engineering, Miami University , Oxford, Ohio
| | - Seth Tomblyn
- 4 KeraNetics, LLC , Winston-Salem, North Carolina
| | - Luke Burnett
- 4 KeraNetics, LLC , Winston-Salem, North Carolina
| | - George J Christ
- 1 Biomedical Engineering Department, University of Virginia , Charlottesville, Virginia.,2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina.,6 Orthopaedics Department, University of Virginia , Charlottesville, Virginia
| |
Collapse
|
45
|
Leijs MJC, van Buul GM, Verhaar JAN, Hoogduijn MJ, Bos PK, van Osch GJVM. Pre-Treatment of Human Mesenchymal Stem Cells With Inflammatory Factors or Hypoxia Does Not Influence Migration to Osteoarthritic Cartilage and Synovium. Am J Sports Med 2017; 45:1151-1161. [PMID: 28114800 DOI: 10.1177/0363546516682710] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are promising candidates as a cell-based therapy for osteoarthritis (OA), although current results are modest. Pre-treatment of MSCs before application might improve their therapeutic efficacy. HYPOTHESIS Pre-treatment of MSCs with inflammatory factors or hypoxia will improve their migration and adhesion capacities toward OA-affected tissues. STUDY DESIGN Controlled laboratory study. METHODS We used real-time polymerase chain reaction to determine the effects of different fetal calf serum (FCS) batches, platelet lysate (PL), hypoxia, inflammatory factors, factors secreted by OA tissues, and OA synovial fluid (SF) on the expression of 12 genes encoding chemokine or adhesion receptors. Migration of MSCs toward factors secreted by OA tissues was studied in vitro, and attachment of injected MSCs was evaluated in vivo in healthy and OA knees of male Wistar rats. RESULTS Different FCS batches, PL, or hypoxia did not influence the expression of the migration and adhesion receptor genes. Exposure to inflammatory factors altered the expression of CCR1, CCR4, CD44, PDGFRα, and PDGFRβ. MSCs migrated toward factors secreted by OA tissues in vitro. Neither pre-treatment with inflammatory factors nor the presence of OA influenced MSC migration in vitro or adhesion in vivo. CONCLUSION Factors secreted by OA tissues increase MSC migration in vitro. In vivo, no difference in MSC adhesion was found between OA and healthy knees. Pre-treatment with inflammatory factors influenced the expression of migration and adhesion receptors of MSCs but not their migration in vitro or adhesion in vivo. CLINICAL RELEVANCE To improve the therapeutic capacity of intra-articular injection of MSCs, they need to remain intra-articular for a longer period of time. Pre-treatment of MSCs with hypoxia or inflammatory factors did not increase the migration or adhesion capacity of MSCs and will therefore not likely prolong their intra-articular longevity. Alternative approaches to prolong the intra-articular presence of MSCs should be developed to increase the therapeutic effect of MSCs in OA.
Collapse
Affiliation(s)
- Maarten J C Leijs
- Department of Orthopaedics, Erasmus MC Rotterdam, the Netherlands.,Department of Radiology, Erasmus MC Rotterdam, the Netherlands
| | | | - Jan A N Verhaar
- Department of Orthopaedics, Erasmus MC Rotterdam, the Netherlands
| | | | - Pieter K Bos
- Department of Orthopaedics, Erasmus MC Rotterdam, the Netherlands
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC Rotterdam, the Netherlands.,Department of Otorhinolaryngology, Erasmus MC Rotterdam, the Netherlands
| |
Collapse
|
46
|
MacArthur JW, Steele AN, Goldstone AB, Cohen JE, Hiesinger W, Woo YJ. Injectable Bioengineered Hydrogel Therapy in the Treatment of Ischemic Cardiomyopathy. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2017; 19:30. [PMID: 28337717 DOI: 10.1007/s11936-017-0530-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OPINION STATEMENT Over the past two decades, the field of cardiovascular medicine has seen the rapid development of multiple different modalities for the treatment of ischemic myocardial disease. Most research efforts have focused on strategies aimed at coronary revascularization, with significant technological advances made in percutaneous coronary interventions as well as coronary artery bypass graft surgery. However, recent research efforts have shifted towards ways to address the downstream effects of myocardial infarction on both cellular and molecular levels. To this end, the broad application of injectable hydrogel therapy after myocardial infarction has stimulated tremendous interest. In this article, we will review what hydrogels are, how they can be bioengineered in unique ways to optimize therapeutic potential, and how they can be used as part of a treatment strategy after myocardial infarction.
Collapse
Affiliation(s)
- John W MacArthur
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - Amanda N Steele
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - Andrew B Goldstone
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - Jeffrey E Cohen
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA.
| |
Collapse
|
47
|
Henry N, Clouet J, Le Visage C, Weiss P, Gautron E, Renard D, Cordonnier T, Boury F, Humbert B, Terrisse H, Guicheux J, Le Bideau J. Silica nanofibers as a new drug delivery system: a study of the protein–silica interactions. J Mater Chem B 2017; 5:2908-2920. [DOI: 10.1039/c7tb00332c] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drug delivery from silica nanofiber based materials for intervertebral disc regenerative medicine.
Collapse
Affiliation(s)
- Nina Henry
- INSERM
- UMRS 1229
- RMeS “Regenerative Medicine and Skeleton”
- Team STEP “Physiopathology and joint regenerative medicine”
- Nantes
| | - Johann Clouet
- INSERM
- UMRS 1229
- RMeS “Regenerative Medicine and Skeleton”
- Team STEP “Physiopathology and joint regenerative medicine”
- Nantes
| | - Catherine Le Visage
- INSERM
- UMRS 1229
- RMeS “Regenerative Medicine and Skeleton”
- Team STEP “Physiopathology and joint regenerative medicine”
- Nantes
| | - Pierre Weiss
- INSERM
- UMRS 1229
- RMeS “Regenerative Medicine and Skeleton”
- Team STEP “Physiopathology and joint regenerative medicine”
- Nantes
| | - Eric Gautron
- Institut des Matériaux Jean Rouxel (IMN)
- UMR 6502 CNRS – Université de Nantes
- Nantes
- France
| | - Denis Renard
- INRA
- UR 1268 Biopolymères Interactions Assemblages
- F-44300 Nantes
- France
| | | | | | - Bernard Humbert
- Institut des Matériaux Jean Rouxel (IMN)
- UMR 6502 CNRS – Université de Nantes
- Nantes
- France
| | - Hélène Terrisse
- Institut des Matériaux Jean Rouxel (IMN)
- UMR 6502 CNRS – Université de Nantes
- Nantes
- France
| | - Jérôme Guicheux
- INSERM
- UMRS 1229
- RMeS “Regenerative Medicine and Skeleton”
- Team STEP “Physiopathology and joint regenerative medicine”
- Nantes
| | - Jean Le Bideau
- Institut des Matériaux Jean Rouxel (IMN)
- UMR 6502 CNRS – Université de Nantes
- Nantes
- France
| |
Collapse
|
48
|
Karpov AA, Udalova DV, Pliss MG, Galagudza MM. Can the outcomes of mesenchymal stem cell-based therapy for myocardial infarction be improved? Providing weapons and armour to cells. Cell Prolif 2016; 50. [PMID: 27878916 DOI: 10.1111/cpr.12316] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/12/2016] [Indexed: 02/06/2023] Open
Abstract
Use of mesenchymal stem cell (MSC) transplantation after myocardial infarction (MI) has been found to have infarct-limiting effects in numerous experimental and clinical studies. However, recent meta-analyses of randomized clinical trials on MSC-based MI therapy have highlighted the need for improving its efficacy. There are two principal approaches for increasing therapeutic effect of MSCs: (i) preventing massive MSC death in ischaemic tissue and (ii) increasing production of cardioreparative growth factors and cytokines with transplanted MSCs. In this review, we aim to integrate our current understanding of genetic approaches that are used for modification of MSCs to enable their improved survival, engraftment, integration, proliferation and differentiation in the ischaemic heart. Genetic modification of MSCs resulting in increased secretion of paracrine factors has also been discussed. In addition, data on MSC preconditioning with physical, chemical and pharmacological factors prior to transplantation are summarized. MSC seeding on three-dimensional polymeric scaffolds facilitates formation of both intercellular connections and contacts between cells and the extracellular matrix, thereby enhancing cell viability and function. Use of genetic and non-genetic approaches to modify MSC function holds great promise for regenerative therapy of myocardial ischaemic injury.
Collapse
Affiliation(s)
- Andrey A Karpov
- Institute of Experimental Medicine, Federal Almazov North-West Medical Research Centre, St Petersburg, Russia.,Department of Pathophysiology, First Pavlov State Medical University of Saint Petersburg, St Petersburg, Russia
| | - Daria V Udalova
- Institute of Experimental Medicine, Federal Almazov North-West Medical Research Centre, St Petersburg, Russia
| | - Michael G Pliss
- Institute of Experimental Medicine, Federal Almazov North-West Medical Research Centre, St Petersburg, Russia
| | - Michael M Galagudza
- Institute of Experimental Medicine, Federal Almazov North-West Medical Research Centre, St Petersburg, Russia.,ITMO University, St Petersburg, Russia
| |
Collapse
|
49
|
Moussa L, Pattappa G, Doix B, Benselama SL, Demarquay C, Benderitter M, Sémont A, Tamarat R, Guicheux J, Weiss P, Réthoré G, Mathieu N. A biomaterial-assisted mesenchymal stromal cell therapy alleviates colonic radiation-induced damage. Biomaterials 2016; 115:40-52. [PMID: 27886554 DOI: 10.1016/j.biomaterials.2016.11.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/14/2016] [Accepted: 11/14/2016] [Indexed: 01/06/2023]
Abstract
Healthy tissues surrounding abdomino-pelvic tumours can be impaired by radiotherapy, leading to chronic gastrointestinal complications with substantial mortality. Adipose-derived Mesenchymal Stromal Cells (Ad-MSCs) represent a promising strategy to reduce intestinal lesions. However, systemic administration of Ad-MSCs results in low cell engraftment within the injured tissue. Biomaterials, able to encapsulate and withstand Ad-MSCs, can overcome these limitations. A silanized hydroxypropylmethyl cellulose (Si-HPMC) hydrogel has been designed and characterized for injectable cell delivery using the operative catheter of a colonoscope. We demonstrated that hydrogel loaded-Ad-MSCs were viable, able to secrete trophic factors and responsive to the inflammatory environment. In a rat model of radiation-induced severe colonic damage, Ad-MSC + Si-HPMC improve colonic epithelial structure and hyperpermeability compared with Ad-MSCs injected intravenously or locally. This therapeutic benefit is associated with greater engraftment of Si-HPMC-embedded Ad-MSCs in the irradiated colonic mucosa. Moreover, macrophage infiltration near the injection site was less pronounced when Ad-MSCs were embedded in the hydrogel. Si-HPMC induces modulation of chemoattractant secretion by Ad-MSCs that could contribute to the decrease in macrophage infiltrate. Si-HPMC is suitable for cell delivery by colonoscopy and induces protection of Ad-MSCs in the tissue potentiating their therapeutic effect and could be proposed to patients suffering from colon diseases.
Collapse
Affiliation(s)
- Lara Moussa
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Recherche en Régénération des tissus sains Irradiés (LR2I), 31 Avenue de la Division Leclerc, 92262 Fontenay-aux-Roses, France; INSERM, Institut National de la Santé et de la Recherche Médicale, UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France; Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France
| | - Girish Pattappa
- INSERM, Institut National de la Santé et de la Recherche Médicale, UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France; Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France
| | - Bastien Doix
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Recherche en Régénération des tissus sains Irradiés (LR2I), 31 Avenue de la Division Leclerc, 92262 Fontenay-aux-Roses, France
| | - Sarra-Louiza Benselama
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Recherche en Régénération des tissus sains Irradiés (LR2I), 31 Avenue de la Division Leclerc, 92262 Fontenay-aux-Roses, France
| | - Christelle Demarquay
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Recherche en Régénération des tissus sains Irradiés (LR2I), 31 Avenue de la Division Leclerc, 92262 Fontenay-aux-Roses, France
| | - Marc Benderitter
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Recherche en Régénération des tissus sains Irradiés (LR2I), 31 Avenue de la Division Leclerc, 92262 Fontenay-aux-Roses, France
| | - Alexandra Sémont
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Recherche en Régénération des tissus sains Irradiés (LR2I), 31 Avenue de la Division Leclerc, 92262 Fontenay-aux-Roses, France
| | - Radia Tamarat
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Recherche en Régénération des tissus sains Irradiés (LR2I), 31 Avenue de la Division Leclerc, 92262 Fontenay-aux-Roses, France
| | - Jérôme Guicheux
- INSERM, Institut National de la Santé et de la Recherche Médicale, UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France; Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France; Centre Hospitalier Universitaire de Nantes, Pôle Hospitalo-Universitaire 4 (OTONN), 1 Place Alexis Ricordeau, 44042 Nantes, France
| | - Pierre Weiss
- INSERM, Institut National de la Santé et de la Recherche Médicale, UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France; Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France; Centre Hospitalier Universitaire de Nantes, Pôle Hospitalo-Universitaire 4 (OTONN), 1 Place Alexis Ricordeau, 44042 Nantes, France
| | - Gildas Réthoré
- INSERM, Institut National de la Santé et de la Recherche Médicale, UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France; Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire (LIOAD), Faculté de Chirurgie Dentaire, 1 Place Alexis Ricordeau, 44042 Nantes, France; Centre Hospitalier Universitaire de Nantes, Pôle Hospitalo-Universitaire 4 (OTONN), 1 Place Alexis Ricordeau, 44042 Nantes, France
| | - Noëlle Mathieu
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Recherche en Régénération des tissus sains Irradiés (LR2I), 31 Avenue de la Division Leclerc, 92262 Fontenay-aux-Roses, France.
| |
Collapse
|
50
|
Yang SL, Tang KQ, Tao JJ, Wan AH, Lin YD, Nan SL, Guo QK, Shen ZY, Hu B. Delivery of CD151 by Ultrasound Microbubbles in Rabbit Myocardial Infarction. Cardiology 2016; 135:221-227. [PMID: 27522674 DOI: 10.1159/000446639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/04/2016] [Indexed: 11/19/2022]
Abstract
OBJECTIVES We aimed to evaluate whether ultrasound (US) and microbubble-mediated delivery of Cluster of Differentiation 151 (CD151) could enhance the therapeutic effects of CD151 on myocardial infarction (MI). METHODS A rabbit model of MI was established by a modified Fujita method. Then, 50 MI rabbits were randomly divided into 5 groups, including G1 (CD151 plasmid and physiological saline in the presence of US); G2 (CD151 and Sonovue in the presence of US); G3 (CD151 and Sonovue in the absence of US); G4 (Sonovue in the absence of US), and a control group (physiological saline in the absence of US). After 14 days of treatment, the expression of CD151 was detected by Western blot. Besides, vessel density of peri-infarcted myocardium was measured by immunohistochemistry, and cardiac function was analyzed by echocardiography. RESULTS The rabbit model of MI was established successfully. CD151 injection increased the expression of CD151 and microvessel density in the myocardium of MI rabbits. Heart function was significantly improved by CD151, which exhibited increased left ventricular ejection fraction, left ventricular fractional shortening and a reduced Tei index. Besides, US Sonovue significantly increased the expression efficiency of CD151. CONCLUSION US microbubble was an effective vector for CD151 delivery. CD151 might be an effective therapeutic target for MI.
Collapse
Affiliation(s)
- Shao-Ling Yang
- Ultrasound in Medicine Departments, Shanghai Fengxian Central Hospital/Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|