Academic literature on the topic 'Adipose-derived stem cells, regenerative medicine'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'Adipose-derived stem cells, regenerative medicine.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "Adipose-derived stem cells, regenerative medicine"

1

Gimble, Jeffrey M., Adam J. Katz, and Bruce A. Bunnell. "Adipose-Derived Stem Cells for Regenerative Medicine." Circulation Research 100, no. 9 (May 11, 2007): 1249–60. http://dx.doi.org/10.1161/01.res.0000265074.83288.09.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Lina, Yani, and Andi Wijaya. "Adipose-Derived Stem Cells for Future Regenerative System Medicine." Indonesian Biomedical Journal 4, no. 2 (August 1, 2012): 59. http://dx.doi.org/10.18585/inabj.v4i2.164.

Full text
Abstract:
BACKGROUND: The potential use of stem cell-based therapies for repair and regeneration of various tissues and organs offers a paradigm shift that may provide alternative therapeutic solutions for a number of disease. Despite the advances, the availability of stem cells remaining a challenge for both scientist and clinicians in pursuing regenerative medicine. CONTENT: Subcutaneous human adipose tissue is an abundant and accessible cell source for applications in tissue engineering and regenerative medicine. Routinely, the adipose issue is digested with collagenase or related lytic enzymes to release a heterogeneous population for stromal vascular fraction (SVF) cells. The SVF cells can be used directly or can be cultured in plastic ware for selection and expansion of an adherent population known as adipose-derived stromal/stem cells (ASCs). Their potential in the ability to differentiate into adipogenic, osteogenic, chondrogenic and other mesenchymal lineages, as well in their other clinically useful properties, includes stimulation of angiogenesis and suppression of inflammation.SUMMARY: Adipose tissue is now recognized as an accessible, abundant and reliable site for the isolation of adult stem cels suitable for the application of tissue engineering and regenerative medicine applications. The past decade has witnessed an explosion of preclinical data relating to the isolation, characterization, cryopreservation, differentiation, and transplantation of freshly isolated stromal vascular fraction cells and adherent, culture-expanded, adipose-derived stromal/stem cells in vitro and in animal models.KEYWORDS: adipose tissue, adult stem cells, regenerative medicine, mesenchymal stem cells
APA, Harvard, Vancouver, ISO, and other styles
3

Skubis, Aleksandra, Bartosz Sikora, Nikola Zmarzły, Emilia Wojdas, and Urszula Mazurek. "Adipose-derived stem cells: a review of osteogenesis differentiation." Folia Biologica et Oecologica 12 (December 7, 2016): 38–47. http://dx.doi.org/10.1515/fobio-2016-0004.

Full text
Abstract:
This review article provides an overview on adipose-derived stem cells (ADSCs) for implications in bone tissue regeneration. Firstly this article focuses on mesenchymal stem cells (MSCs) which are object of interest in regenerative medicine. Stem cells have unlimited potential for self-renewal and develop into various cell types. They are used for many therapies such as bone tissue regeneration. Adipose tissue is one of the main sources of mesenchymal stem cells (MSCs). Regenerative medicine intends to differentiate ADSC along specific lineage pathways to effect repair of damaged or failing organs. For further clinical applications it is necessary to understand mechanisms involved in ADSCs proliferation and differentiation. Second part of manuscript based on osteogenesis differentiation of stem cells. Bones are highly regenerative organs but there are still many problems with therapy of large bone defects. Sometimes there is necessary to make a replacement or expansion new bone tissue. Stem cells might be a good solution for this especially ADSCs which manage differentiate into osteoblast in in vitro and in vivo conditions.
APA, Harvard, Vancouver, ISO, and other styles
4

Frese, Laura, Petra E. Dijkman, and Simon P. Hoerstrup. "Adipose Tissue-Derived Stem Cells in Regenerative Medicine." Transfusion Medicine and Hemotherapy 43, no. 4 (2016): 268–74. http://dx.doi.org/10.1159/000448180.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Konno, Masamitsu, Atsushi Hamabe, Shinichiro Hasegawa, Hisataka Ogawa, Takahito Fukusumi, Shimpei Nishikawa, Katsuya Ohta, et al. "Adipose-derived mesenchymal stem cells and regenerative medicine." Development, Growth & Differentiation 55, no. 3 (March 3, 2013): 309–18. http://dx.doi.org/10.1111/dgd.12049.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Alt, Eckhard U., Glenn Winnier, Alexander Haenel, Ralf Rothoerl, Oender Solakoglu, Christopher Alt, and Christoph Schmitz. "Towards a Comprehensive Understanding of UA-ADRCs (Uncultured, Autologous, Fresh, Unmodified, Adipose Derived Regenerative Cells, Isolated at Point of Care) in Regenerative Medicine." Cells 9, no. 5 (April 29, 2020): 1097. http://dx.doi.org/10.3390/cells9051097.

Full text
Abstract:
It has become practically impossible to survey the literature on cells derived from adipose tissue for regenerative medicine. The aim of this paper is to provide a comprehensive and translational understanding of the potential of UA-ADRCs (uncultured, unmodified, fresh, autologous adipose derived regenerative cells isolated at the point of care) and its application in regenerative medicine. We provide profound basic and clinical evidence demonstrating that tissue regeneration with UA-ADRCs is safe and effective. ADRCs are neither ‘fat stem cells’ nor could they exclusively be isolated from adipose tissue. ADRCs contain the same adult stem cells ubiquitously present in the walls of blood vessels that are able to differentiate into cells of all three germ layers. Of note, the specific isolation procedure used has a significant impact on the number and viability of cells and hence on safety and efficacy of UA-ADRCs. Furthermore, there is no need to specifically isolate and separate stem cells from the initial mixture of progenitor and stem cells found in ADRCs. Most importantly, UA-ADRCs have the physiological capacity to adequately regenerate tissue without need for more than minimally manipulating, stimulating and/or (genetically) reprogramming the cells for a broad range of clinical applications. Tissue regeneration with UA-ADRCs fulfills the criteria of homologous use as defined by the regulatory authorities.
APA, Harvard, Vancouver, ISO, and other styles
7

Sultanova, A. S., O. Ya Bespalova, and O. Yu Galkin. "Stromal-vascular fraction of adipose tissue as an alternative source of cellular material for regenerative medicine." Ukrainian Biochemical Journal 93, no. 1 (February 22, 2021): 40–50. http://dx.doi.org/10.15407/ubj93.01.040.

Full text
Abstract:
Adipose tissue is the most convenient source of cellular material for regenerative medicine as it can be obtained in significant quantities via cosmetic liposuction, lipoaspiration of subcutaneous fat or by excision of fat deposits. Adipose tissue consists of adipocytes and cells, which are the part of the stromal-vascular fraction (SVF). Different cell populations can be isolated from SVF, among which the population of adipose tissue stem cells (adipose-derived stem cells, ADSC) is especially important for regenerative medicine. SVF can be obtained relatively easily from adipose tissue (adipose tissue is an alternative to bone marrow in terms of being a source of stem cells) and used to treat various pathologies. Recent studies show that SVF not only has a therapeutic effect similar to that of ADSC, but in some cases is even more effective. The article provides the analysis of the main methods of SVF obtainment, characteristics of SVF cellular composition, its potential for use in clinical medicine and its main advantages over other sources of cellular material, including­ ADSC cultured in vitro, for regenerative medicine. Keywords: adipocytes, adipose-derived stem cells, regenerative medicine, stromal-vascular fraction
APA, Harvard, Vancouver, ISO, and other styles
8

Levi, Benjamin, and Michael T. Longaker. "Concise Review: Adipose-Derived Stromal Cells for Skeletal Regenerative Medicine." STEM CELLS 29, no. 4 (April 2011): 576–82. http://dx.doi.org/10.1002/stem.612.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Zuk, Patricia. "Adipose-Derived Stem Cells in Tissue Regeneration: A Review." ISRN Stem Cells 2013 (February 14, 2013): 1–35. http://dx.doi.org/10.1155/2013/713959.

Full text
Abstract:
In 2001, researchers at the University of California, Los Angeles, described the isolation of a new population of adult stem cells from liposuctioned adipose tissue. These stem cells, now known as adipose-derived stem cells or ADSCs, have gone on to become one of the most popular adult stem cells populations in the fields of stem cell research and regenerative medicine. As of today, thousands of research and clinical articles have been published using ASCs, describing their possible pluripotency in vitro, their uses in regenerative animal models, and their application to the clinic. This paper outlines the progress made in the ASC field since their initial description in 2001, describing their mesodermal, ectodermal, and endodermal potentials both in vitro and in vivo, their use in mediating inflammation and vascularization during tissue regeneration, and their potential for reprogramming into induced pluripotent cells.
APA, Harvard, Vancouver, ISO, and other styles
10

Yeh, Da-Chuan, Tzu-Min Chan, Horng-Jyh Harn, Tzyy-Wen Chiou, Hsin-Shui Chen, Zung-Sheng Lin, and Shinn-Zong Lin. "Adipose Tissue-Derived Stem Cells in Neural Regenerative Medicine." Cell Transplantation 24, no. 3 (March 2015): 487–92. http://dx.doi.org/10.3727/096368915x686940.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "Adipose-derived stem cells, regenerative medicine"

1

Srivastava, Sapna. "The potential of human adipose derived stem cells for myocardial regenerative therapy." Thesis, McGill University, 2010. http://digitool.Library.McGill.CA:80/R/?func=dbin-jump-full&object_id=95088.

Full text
Abstract:
Background: Cell therapy using Human Bone Marrow Stem Cells (HBMSCs) has been shown to improve heart function after a myocardial infarction. The harvesting technique involved with bone marrow stem cells is invasive and yields a low cell number. There is now an increasing interest in Human Adipose Derived Stem Cells (HADSCs) as they are abundant and readily accessible from liposuction material. The present study was undertaken to investigate if HADSCs are superior than HBMSCs in myocardial regenerative therapy. Results: Both HADSCs and HBMSCs proliferated in a time dependent manner, however, the proliferative ability of HADSC was greater than HBMSCs. In addition, both cells differentiated to the osteoblast lineage confirming their multipotency when treated with induction medium. Furthermore, treatment of both cells with 5-AC resulted in positive immunostaining of cardiac markers, troponinI and connexin 43, however the expression of these markers was enhanced in HADSCs. This was further confirmed by western blot analysis, however 5-AC treatment did not exhibit cell contraction or multinucleation. In addition, these results were further confirmed by our in vivo study. Both cells were injected in the heart of a rat model of myocardial infarction and was monitored for ejection fraction (EF) and fractional shortening (FS) for 24 hours, 3 weeks and 6 weeks post-surgery. The cardiac function of the rats treated with stem cells was improved as demonstrated by increase in EF and FS, however, a greater improvement was seen with HADSCs compared with HBMSCs. This notion is further substantiated by our studies on left ventricular infarct size measurement, showing that HADSC are more potent in reduction of the infarct compared to HBMSCs. Conclusion: The data suggest that HADSCs may prove to be a more ideal alternative for regenerative therapy in the future.
La thérapie cellulaire à l'aide de cellules souches humaines de la moelle osseuses (CSHMOs) a été démontré d'améliorer la fonction cardiaque après un infarctus du myocarde. La technique de récolte des CSHMOs est pourtant invasive et donne un nombre de cellules viables faible. Il y a maintenant un intérêt croissant dans les cellules souches humaines dérivés du tissu adipeux (CSHTAs), car elles sont abondantes et facilement accessibles à partir des amas de graisses provenant des chirurgies de liposuccion. La présente étude a été menée pour vérifier si les CSHTAs sont supérieures aux CSHMOs dans la thérapie régénératrice du myocarde. Résultats: Les CSHTAs ainsi que les CSHMOs ont proliféré dans une manière temps dépendante, cependant, la capacité proliférative des CSHTAs était supérieure à celle des CSHMOs. De plus, les deux types de cellules souches ce sont différenciées en lignée ostéoblastique, affirmant leur capacité multipotent lorsqu'elles sont traitées avec le milieu d'induction. En outre, le traitement des deux types de cellules souches avec le 5-AC a entraîné l'immunomarquage positif de troponin I et de connexine 43, marqueurs cardiaques, cependant l'expression de ces marqueurs était plus robuste dans les CSHTAs. Cela a été confirmé par analyse d'immunobuvardage de type Western, cependant les cellules traité au 5-AC ne présentait pas de contraction des cellules ou le développement de plusieurs noyaux. En plus, ces résultats ont été confirmés par nos études in vivo. Les deux types de cellules ont été injectées dans le cœur d'un modèle de rat d'infarctus du myocarde et a été suivie pour la fraction d'éjection (FE) et la fraction de raccourcissement (FR) pour 24 heures, 3 semaines et 6 semaines post-chirurgie. La fonction cardiaque des rats traités avec les cellules souches a été améliorée, fait démontré par l'augmentation de l'FE et le FR, cependant, une plus grande amélioration de ce
APA, Harvard, Vancouver, ISO, and other styles
2

Francis, Michael. "RECAPITULATING OSTEOBLASTOGENESIS WITH ELECTROSPUN FIBRINOGEN NANOFIBERS AND ADIPOSE STEM CELLS AND ELECTROSPINNING ADIPOSE TISSUE-DERIVED BASEMENT MEMBRANE." VCU Scholars Compass, 2010. http://scholarscompass.vcu.edu/etd/2025.

Full text
Abstract:
To repair, replace, or regenerate damaged or diseased tissue has been a long-standing, albeit elusive, goal in medical research. Here, we characterize patient-derivable mesenchymal stem cell types, termed adipose-derived stem cells (ASCs). These cells, which can be derived from liposuction fat and lipoaspirate saline, are sources for patient-derivable extracellular matrix (ECM), fibrinogen (Fg) and adipose tissue extracellular matrix, and may prove useful for synthesizing new bone tissue analogues in vitro. Traditionally and rapidly isolated ASCs were thoroughly characterized as multipotent, having osteogenic, adipogenic, and chondrogenic differentiation potential, and they exhibited comparable proliferative lifespans. These ASCs also shared an indistinguishable immunophenotype when compared to bone marrow-derived mesenchymal stem cells, suggesting that these cells are an excellent source for bone following tissue engineering experimentation. In order to synthesize bone ex-vivo, electrospun scaffolds of fibrinogen (Fg), polydioxanone (PDO), and Fg:PDO blends were seeded with early passage ASCs, fibroblasts, or osteosarcoma cells and were maintained for 21 days in osteogenic or regular growth media. Constructs were analyzed both histologically and molecularly for evidence of osteoblastogenesis. Using SEM, the appearance of regular, mineralized-appearing structures were found in osteogenic-induced ASC seeded scaffolds beyond 14 days, only in the scaffolds containing Fg. Further, at 21 days of culture, Fg scaffolds with ASCs in osteogenic media became hard and brittle. Robust new collagen synthesis and matrix remodeling were observed on all Fg scaffolds, the levels of which were elevated over time. Pronounced mineralization was found throughout bone-induced ASC scaffolds, while control scaffolds (BJ foreskin fibroblasts) showed no mineral deposition (although they did demonstrate excellent cellularity). Analysis of gene expression (qRT-PCR) indicated that electrospun Fg supported osteoblastogenesis through the upregulation of alkaline phosphatase and osteocalcin gene expression. To confirm our gene expression results, osteogenic-induced ASCs on Fg scaffolds were also shown to secrete osteocalcin in the extracellular matrix, a key marker in osteoblastogenesis. Thus, electrospun Fg is an excellent material for ASC growth, proliferation, and osteogenic differentiation, providing an ideal system for furthering basic bone model-based research and for advancing regenerative medicine. In addition to establishing Fg as a source of scaffolding, we developed and characterized a novel method for isolating and subsequently electrospinning adipose tissue matrix. Because adipose ECM contains many primordial matrix proteins important for embryonic development and regeneration (such as laminin, type IV collagen, and fibronectin), adipose ECM may prove to be an autologous tissue engineering matrix and stem cell culture substrate. We show here that adipose tissue ECM can, in fact, be electrospun into a nanofiberous mesh, histologically shown to contain connective tissue, collagens, elastic fibers/elastin, proteoglycans, and glycoproteins in the newly synthesized matrix. We also show that this novel electrospun adipose tissue scaffold is capable of supporting stem cell growth. Taken together, experiments using ASCs cultured on extracellular matrices of electrospun Fg or adipose ECM present an excellent framework for future advances in regenerative medicine therapeutics and research.
APA, Harvard, Vancouver, ISO, and other styles
3

Sutha, Ken. "Osteoinductive material derived from differentiating embryonic stem cells." Diss., Georgia Institute of Technology, 2012. http://hdl.handle.net/1853/51722.

Full text
Abstract:
The loss of regenerative capacity of bone, from fetal to adult to aged animals, has been attributed not only to a decline in the function of cells involved in bone formation but also to alterations in the bone microenvironment that occur through development and aging, including extracellular matrix (ECM) composition and growth/trophic factor content. In the development of novel treatments for bone repair, one potential therapeutic goal is the restoration of a more regenerative microenvironment, as found during embryonic development. One approach to creating such a microenvironment is through the use of stem cells. In addition to serving as a differentiated cell source, pluripotent stem cells, such as embryonic stem cells (ESCs), may possess the unique potential to modulate tissue environments via local production of ECM and growth factors. ESC-produced factors may be harnessed and delivered to promote functional tissue regeneration. Such an approach to generate a naturally derived, acelluar therapy has been employed successfully to deliver osteoinductive factors found within adult bone, in the form of demineralized bone matrix (DBM), but the development of treatments derived instead from developing, more regenerative tissues or cells remains attractive. Furthermore, the derivation of regenerative materials from an ESC source also presents the added benefit of eliminating donor to donor variability of adult, cadaveric tissue derived materials, such as DBM. Thus, the objective of this project was to examine the osteoinductive potential harbored within the embryonic microenvironment, in vitro and in vivo. The osteogenic differentiation of mouse ESCs as embryoid bodies (EBs) was evaluated in response to phosphate treatment, in vitro, including osteoinductive growth factor production. The osteoinductivity of EB-derived material (EBM) was then compared to that of adult tissue-derived DBM, in vivo. Phosphate treatment enhanced osteogenic differentiation of EBs. EBM derived from phosphate treated EBs retained bioactive, osteoinductive factors and induced new bone formation, demonstrating that the microenvironment within osteogenic EBs can be harnessed in an acellular material to yield in vivo osteoinductivity. This work not only provides new insights into the dynamic microenvironments of differentiating stem cells but also establishes an approach for the development of an ESC-derived, tissue specific therapy.
APA, Harvard, Vancouver, ISO, and other styles
4

Lazin, Jamie Jonas. "The effect of age and sex on the number and osteogenic differentiation potential of adipose-derived mesenchymal stem cells." Thesis, Georgia Institute of Technology, 2010. http://hdl.handle.net/1853/34696.

Full text
Abstract:
It has been shown that stem cells exist within adult adipose tissue. These stem cells are named adipose-derived mesenchymal stem cells (ASCs), are derived from the mesoderm, and can differentiate into a number of cells including osteoblasts, chondrocytes, and adipocytes. However, before these cells can be used clinically it is important that we understand how factors like age, sex, and ethnicity affect ASC number and potential. Additionally, since men and women vary in their distribution of adipose tissue, it will be important to see if the ideal source of ASCs is different for each sex. The goal of this study was to assess how age and sex affects ASCs. We used flow cytometry to investigate how age and sex affected the number of ASCs in adipose tissue. Additionally, we plated these cells in culture and treated them with an osteogenic media (OM) with the intention of pushing them towards osteoblast differentiation. The purpose of this was to see if age or sex affected the potential of the ASCs to undergo osteogenesis in culture. For this study we used real-time PCR and biochemical assays to look at markers of early and late osteogenic differentiation. Finally, we used immunohistochemistry to demonstrate where in adipose tissue the CD73 and CD271 positive cell population exists. It is our hope that this work will shed light on how age and sex affect ASCs so that clinicians can optimize their ASC harvest depending on the patient's physiology.
APA, Harvard, Vancouver, ISO, and other styles
5

Nair, Rekha. "Acellular matrices derived from differentiating embryonic stem cells." Diss., Georgia Institute of Technology, 2009. http://hdl.handle.net/1853/37170.

Full text
Abstract:
Embryonic stem cells (ESCs) can differentiate into all somatic cells, and as such, are a promising cell source for therapeutic applications. In vitro, ESCs spontaneously differentiate via the aggregation of cells into embryoid bodies (EBs), which recapitulate aspects of early embryogenesis and harbor a unique reservoir of cues critical for tissue formation and morphogenesis. Embryonic healing responses employ similar intrinsic machinery used for tissue development, and these morphogenic cues may be captured within the EB microenvironment. Recent studies have shown that when injected into injury or defect models in vivo, ESCs synthesize and secrete extracellular factors that ultimately contribute to repair, suggesting that these molecules may be as important for regenerative therapies as functional differentiation of the cells. The overall objective of this project was to develop novel acellular matrices derived from differentiating ESCs undergoing morphogenesis. The central hypothesis was that embryonic matrices contain complex mixtures of extracellular factors that, when isolated, retain bioactivity and enhance wound healing in an adult environment. The overall objective was accomplished by: (1) investigating the production of extracellular matrix (ECM) by differentiating ESCs as a function of differentiation time; (2) assessing the ability of solvents to efficiently decellularize EBs; and (3) evaluating the healing response elicited by acellular matrices derived from EBs in a murine dermal wound healing model. Endogenous ECM synthesis by EBs varied with time and was associated with specific differentiation events. Novel techniques were developed to effectively remove cell components from EBs in order to extract complex, bioactive acellular matrices. EB-derived acellular matrices significantly enhanced the healing of excisional dermal wounds in mice, indicating the potency of extracellular factors synthesized by ESCs. All together, these studies demonstrate that acellular matrices derived from ESCs retain morphogenic factors capable of influencing tissue repair. In addition, this work lays the foundation for future studies to further examine the functional role of endogenous matrix molecules on ESC differentiation and to evaluate the utility of a stem cell-derived matrix for a variety of regenerative medicine applications.
APA, Harvard, Vancouver, ISO, and other styles
6

Yasin, Mohammed. "Non-regenerative benefits of adult bone marrow derived stem cells for myocardial protection." Thesis, Queen Mary, University of London, 2013. http://qmro.qmul.ac.uk/xmlui/handle/123456789/8701.

Full text
Abstract:
Ischaemic heart disease is the most common cause of mortality in the western hemisphere and it is rapidly becoming the leading cause of death globally. Moreover, therapeutic interventions by cardiologists and cardiac surgeons frequently subject the heart to acute I/R injury, which in itself can cause mortality. Recent investigations of adult stem cells have primarily focused on their regenerative potential for chronic ischaemic heart disease. In this thesis, I have investigated the hypothesis that adult bone marrow derived stem cells are cardioprotective in acute regional myocardial I/R injury. In a rat model of left anterior descending coronary artery (LAD) reversible occlusion and reperfusion, I demonstrate that an intravenous bolus of adult bone marrow derived (1) bone marrow mononuclear (BMNNC) and (2) mesenchymal stem cells (MSC) upon reperfusion can attenuate infarct size. This effect is comparable to ischaemic preconditioning (IPC), which is the gold standard for cardioprotection. Next, I demonstrated the mechanisms for adult stem cell cardioprotection are principally anti-apoptotic and depend upon stem cell secreted factors to (1) activate phosphatidylinositide 3-kinase (PI3)/Akt cell survival kinase-signaling pathway (2) inhibit glycogen synthase kinase-3β (3) inhibit p38MAPK (4) inhibit nuclear translocation of p65NF-κB. 7 Proteomic analysis of myocardium subjected to I/R and treated with either BMMNC or BMMNC derived supernatant (BMS) upon reperfusion demonstrated higher expression of a whole host of pro-survival proteins. These were notably (1) 14-3-3-ε protein (2) anti-oxidant peroxiredoxin-6 (3) heat shock protein (HSP) αB-crystallin, HSP72, HSP tumour necrosis factor receptor-1 associated protein, and HSP ischaemia responsive protein-94 (4) glycolytic protein glyceraldehyde-3-phosphate dehydrogenase (5) mitochondrial aconitase and mitochondrial voltage-dependent anionselective channel protein-1. Thereafter, I investigated the mobilization of endogenous bone marrow stem cells and trafficking to the ischaemic myocardium by stromal cell derived factor-1 (SDF-1) /chemokine, receptor type 4 (CXCR4) signaling. I demonstrate high up-regulated expression of CXCR4 and CD26 in BMMNC following IPC, which might have a role in IPC-mediated cardioprotection. Finally, and in concordance with this finding I demonstrate that both IPC and an exogenous MSC bolus upon reperfusion can synergize to abolish acute myocardial I/R injury.
APA, Harvard, Vancouver, ISO, and other styles
7

Arrigoni, E. "ADIPOSE-DERIVED STEM CELLS (ASCS) FOR FUTURE CELLULAR THERAPIES IN MUSCLE-SKELETAL TISSUES REGENERATION." Doctoral thesis, Università degli Studi di Milano, 2012. http://hdl.handle.net/2434/170261.

Full text
Abstract:
Every year several patients have to deal with bone tissue loss due to trauma or diseases. Bone tissue engineering aims to restore or repair musculoskeletal disorders through the development of bio-substitutes that require the use of cells and scaffolds which should possess both adequate mechanical properties and interconnecting pores to allow cellular infiltration, graft integration and vascularization. The ideal cell for tissue engineering should possess a potential plasticity with the ability to functionally repair the damaged tissue, and it should be available in large amount. Mesenchymal stem cells (MSCs) are present in many adult tissues, and adipose tissue represents an attractive source of MSCs for researchers and clinicians of nearly all medical specialties. Adipose-derived stem cells (ASCs) are similar to MSCs isolated from bone marrow, placenta, and umbilical cord blood in morphology, immunophenotype, and differentiation ability, and they represent a promising approach of bone regeneration. Additional features of ASCs are their immunoregolatory and anti-inflammatory properties both in vivo and in vitro and their low immunogenicity. Since several years our laboratory is studying mesenchymal stem cells isolated from human and animal adipose tissues. Human ASCs (hASCs) have been characterized by their immunophenotype, their self-renewal potential, and they have been induced to differentiate towards adipogenic, osteogenic and chondrogenic lineages. The ability of hASCs to grow in the presence of several scaffolds has also been tested. hASCs adhered to the surface of tested biomaterials, filling the pores and forming a 3D web-like structure, allowing these progenitor cells to osteo-differentiate more efficiently respect to cells maintained on polystyrene. Since our interest was to regenerate muscle-skeletal defects by ASCs in pre-clinical models, we first studied ASCs isolated from adipose tissue of rat (rASCs), rabbit (rbASCs) and pig (pASCs), considered good models in the orthopaedic field. We have shown that animal ASCs behaved similarly to the human ones, and, in collaboration with the Faculty of Veterinary Medicine of University of Milan and the IRCCS Galeazzi Orthopaedic Institute of Milan, we have tested the ability of autologous ASCs to regenerate a full-thickness critical-size bone defect in rabbits. The experimental study was conducted on the tibiae of 12 New Zealand rabbits, and from 6 rabbits out of 12 we have collected adipose tissue from the interscapular region. We have isolated 2.8x105±1.9x105 rbASCs per ml of raw tissue, and after 3-4 days in culture the cells showed the typical fibroblast-like morphology. One week later, all the 6 cellular populations started to steadily proliferate, and they generated fibroblast (CFU-F) and osteoblast (CFU-O) colonies, highlighting the presence of osteogenic progenitors. Indeed, when rbASCs were induced to osteo-differentiate, either after 7 and 14 days, we have observed an up-regulation of specific osteogenic markers, such as alkaline phosphatase (ALP, +28.9%), collagen (+105.9%) and extracellular calcified matrix (+168.1%), compared to undifferentiated cells. In parallel, testing HA, the scaffold selected for the in vivo experiment, we found that rbASCs were osteoinduced; indeed the presence of HA granules increased per se the amount of collagen production (+48.2%). 1.5x106 undifferentiated rbASCs were seeded on custom-made HA disks (8 mm Ø x 4 mm ↕), and the day after, each bioconstruct was implanted into the lesion created in the tibia of each rabbit. We had an additional experimental group of defects where the same number of rbASCs were inserted in the lesion as a semi-liquid suspension; moreover, as controls, we treated 6 lesioned tibia with just the scaffolds, and we left 6 untreated lesioned bone. 8 weeks after surgery animals were sacrificed and the tibia explanted. A macroscopic analysis showed no bone resorption, no abnormal bone callous formation, no fractures, infection or inflammatory reactions, and all the bone defects were completely filled without any significant differences among the four groups. Interestingly, in the presence of scaffold seeded with rbASCs, histology and immunohistochemistry showed a new bone tissue more mature and similar to the native bone. These data have also been confirmed by biomechanical tests: indeed, the mechanical properties of the bone defect treated by rbASCs-HA were improved, suggesting that these constructs bore mechanical loading with an increase in stiffness of 19.8% and in hardness of 31.6% respect to just HA treated group, indicating that the bioconstructs made out of autologous rbASCs and hydroxyapatite might ameliorate the treatment for large bone defects. We would suggest the use of ASCs as a safe cellular therapy in future clinical applications where a large bone defect needs to be treated. These promising results on small size animals allow us to plan a new study on large size animals such as minipigs. However, before moving to the clinic, we know that there are several important aspects that need to be faced regarding safeness and the features of the candidate patients: 1. may the “quality” of hASCs be affected by the donor’s physiological or pathological conditions? 2. may the use of pharmacological treatment enhance cellular plasticity of multipotent cells? 3. may the use of immunoselected hASCs ameliorate tissue regeneration in the field of muscle-skeletal? We have addressed some of these aspects, comparing different populations of hASCs from subcutaneous adipose tissue of healthy-young-female donors (hASCs<35 y/o, n=12, mean age 31±4 years, BMI=23.5±1.6), and from middle-age ones (hASCs>45 y/o n=14, mean age 56±7 years, mean BMI=28.4±1.8). The cellular yield of hASCs derived from older donors was 2.5 fold greater than the one of hASCs<35 y/o, whereas hASCs from younger donors were more clonogenic than hASCs isolated from older ones, with an increase of 129%. No significant differences were observed looking at their immunophenotype. When hASCs were induced to differentiate into cells of the adipogenic and osteogenic lineages, the donor’s age did not affect their adipogenic differentiation, whereas the osteogenic one was significantly affected by age both in the absence and in the presence of three-dimensional scaffolds, showing a decreased ALP basal levels of about 10-fold in hASCs>45 y/o respect to hASCs<35 y/o. These results seems to indicate that ASCs from different donors could behave differently. Trying to overcome this aspect we have used different approaches, and we have studied if Reversine, a synthetic purine already known to increase plasticity of terminally differentiated cells, might improve the differentiation ability of hASCs. 72 hours treatment with 50 nM Reversine induced hASCs to differentiate into osteoblast like-cells (+45% of alkaline phosphatase activity), smooth muscle cells (+89% of α-actin expression) and skeletal muscle cells (myotubes formation) compared to control hASCs. Moreover, since it is known that CD34 and L-NGFR positive cells define a subset of high proliferative and multipotent MSCs, we have immunoselected, these progenitor cells from hASC populations. In contrast to the whole population, the immunoseparated fractions maintained their undifferentiate state and their ability to differentiate much longer during culture. We have shown that both CD34+ and L-NGFR+ hASCs can be used as alternative candidates for tissue engineering and regenerative medicine applications. In particular, due to the improved ability of L-NGFR positive cells to adipo- and chondro-differentiate, they appear an ideal tool in reconstructive plastic surgery and cartilage regeneration. From our data, and the ones from researchers in other fields, we believe that in the near future adipose-derived stem cells might be considered a safe tool in regenerative medicine. Furthermore, to improve this “cellular therapy”, we could either pre-treat ASCs with molecules, such as drugs and/or siRNAs known to affect specific differentiation pathways, or by selecting subpopulations of progenitor cells which may be used as allogenic implants. Next step will be to confirm our in vivo data in a large size animal model such as minipig, and then to test if pre-treated cells or selected population might be used in an autologous and allogenic small size animal model.
APA, Harvard, Vancouver, ISO, and other styles
8

GRILLI, FEDERICA. "Controlling the Adipose-derived Stem cell 3D-organization on micrometric PLGA regular scaffolds for cardiac tissue regeneration and repair." Doctoral thesis, Università degli studi di Genova, 2022. http://hdl.handle.net/11567/1091894.

Full text
Abstract:
The deposition of cells at sites of injury is a clinically relevant approach to facilitate local tissue regeneration and repair. However, cell engraftment, retention, and survival are generally modest, requiring the development of novel deposition techniques and biomaterials. Here, a micro-sized polymeric network (microMESH) is investigated as a promising biodegradable scaffold for the engraftment and tissue integration of human Adipose-Derived Stem Cells (hADSCs) to be used for a wide range of injuries, including myocardial infarction. microMESH comprises a regular network of PLGA microfilaments spatially organized to form square openings of 5x5, 10x10 and 20x20 μm2 . microMESH is realized using soft lithographic techniques starting from a master silicon template reproducing the actual geometry of the final PLGA network. After extensive geometrical, physico-chemical, and mechanical characterizations using a broad range of techniques, hADSCs were integrated with microMESH. Cell viability, spatial organization, secretome and stemness were characterized for all three different microMESH configurations and compared to conventional systems, including 2D plastic dishes and collagen layers. Interestingly, when hADSCs were cultured on microMESH they organized in spheroidal-like structures, despite the geometry, maintaining viability over time. This peculiar attitude of the microMESH to form assemblies better represents the human tissue outside the body, compared to 2D monolayer cultures. Additionally, spheroids established an intimate interaction with the microMESH resulting in the scaffold incorporation within the 3D arrangement formed by the cells. On the contrary, hADSCs form only superficial interaction above the flat collagen sheet that is currently used for cell transplantation in animal models of cardiac diseases. Moreover, once the hADSCs are placed on microMESH, the actin cytoskeleton reorganizes to confer a 3D cell shape with multidirectional actin arrangements, forming nonlinear structures and ring structures at the anchorage site to the microMESH, relative to linear filaments when the cells adhered and flattened onto the plastic surface and on top of collagen scaffold. This internal reorganization and the stronger interaction may explain why microMESH scaffold fostered the secretion of biologically active molecules, acting in a paracrine fashion on resident cells, which are expected to accelerate tissue regeneration and repair. Specifically, when hADSCs grew on microMESH we observed a trend for higher production of several factors with specific implications in angiogenesis, stem cell proliferation and expansion, cell survival, inflammation modulation, ECM remodeling, stem cell mobilization, chemotaxis and homing, relative to 2D monolayer conditions. The paracrine effect of hADSCs is scaffold dependent and can be modulated by tailoring the geometrical and mechanical properties of microMESH. Indeed, the 5x5 microMESH showed its contribution in angiogenesis, ECM remodeling and stem cell mobilization from bone marrow into the bloodstream. Indeed, highest amounts of VEGF, TIMP-2 and GCSF, respectively were detected in 5x5 geometry compared to the other conditions. Rather, 10x10 geometry promotes angiogenesis enhancing the VEGF production, stem cell proliferation and survival by raising the Fibroblast Growth Factors family secretion and EGF factor, respectively and favors ECM remodeling increasing the TIMP-2 production compared to other conditions. Lastly, the 20x20 seems to have a more anti-inflammatory role (combination of IL-10 and TGF-β1) and chemotactic function (e.g. RANTES). Finally, in this work, we started to shed new light on the ability of micromMESH geometry to modulate the hADSCs stemness evaluating the expression levels of CD44, CD90 and CD105 markers over time. The proposed microMESH scaffold is expected to provide an effective alternative to more conventional hADSCs transplant techniques.
APA, Harvard, Vancouver, ISO, and other styles
9

Caremoli, F. "PURIFICATION, CHARACTERIZATION AND CULTURE OF ENSHEATHING CELLS FROM HUMAN OLFACTORY MUCOSA BIOPSIES." Doctoral thesis, Università degli Studi di Milano, 2015. http://hdl.handle.net/2434/335140.

Full text
Abstract:
Among all the possible sources of mesenchymal stem cells, adipose tissue and olfactory mucosa have raised great interest and have become some of the most investigated sources. Adipose tissue-derived mesenchymal stem cells and the fat itself as a source of human adipose derived stem cells, represent one of the major fields of research in regenerative medicine. A great advantage is represented by the minimal invasive and high accessibility to adipose tissue and its ready availability. In the present study, hADSCs were isolated from the adipose tissue donated by several patient and have been investigated and characterized through different technical approaches, such as flow cytometry and immunocytochemistry. These hADSCs reproducibly fulfill the general definition of MSCs by both phenotypic and differentiation capabilities criteria, showing also the expression of neural markers, as observed by confocal microscope analysis. Lipoaspirated adipose tissue showed positivity to ß-tubulin III that was also maintained in lipoaspirate-derived hADSCs. A population of stem cells retaining typical characteristics of surface markers of classical adipose tissue stem cells and MSC was obtained when adipose tissue was subjected to culture in vitro, either by processing through centrifugation or by direct plating without enzymatic digestion with collagenase. Flow cytometry analyses showed that hADSCs expressed classical mesenchymal markers such as CD44, CD73, CD90, CD105 and CD166, while endothelial (CD31, CD34, CD144, CD146) and hematopoietic (CD45, CD133) markers were much less represented. Also the ability to give rise to tissue of mesenchymal origins, such as osteoblastic and adipogenic lineages, were present in hADSCs. In addition, the immunofluorescence staining indicated the expression of neural stem markers in hADSCs which consequently co-expressed nestin, β-tubulin III and glial GFAP. We have also characterized human olfactory ensheathing stem cells. Olfactory mucosa is specialized tissue inside the nasal cavity involved in olfactory perception and capable of lifelong regeneration throughout adulthood. Multipotent stem cells obtained from it offer the possibility of promoting regeneration and reconstruction in regenerative medicine, being readily accessible with minimal invasive techniques, capable of expansion in vitro and retaining broadly potent differentiative capacity as stem cell progenitors. Among the several members of the olfactory mucosa, Olfactory Ensheathing Cells (OECs) are well known to be useful in repairing the nervous system. By following our method, cells can be easily isolated and maintained in TCM, and their cultivation in large flasks allowed obtaining rich cultures of OECs in 2 weeks. Cell cycle analysis showed that the majority of cells are in G0/G1 phase, while just a lesser part is in S/G2 phase. In our growth conditions, no chromosomal abnormalities were observed also at high culture passage (p14). Live morphology of obtained cells showed a fibroblast-like phenotype and the immunohistochemical analyses showed the expression of beta-Tubulin III, Vimentin, Nestin, Glial Fibrillary Acidic Protein and Microtubule-Associated Protein 2. By FACS analysis we demonstrated that OECs are positive to typical surface mesenchymal markers (CD44, CD73, CD90, CD105, CD146 and CD166). As expected, some endothelial (CD31, CD34) and hematopoietic (CD45) markers were very few represented, while some others (CD56, CD144, CD146, CD133) are partially found. These cells also express genes that constitute the core circuitry of self-renewal such as SOX2, NANOG and OCT4 and the stemness marker CD133. OECs incubated with serum-free medium, normally used for the formation of neurospheres, spontaneously formed large spheroids reaching a mean diameter of 100 μm in 10 days of culture. Immunofluorescence of specific proteins showed that spheres were positive to markers such as Nestin, Vimentin, TUJ-1, MAP2 and GFAP. In conclusion, our method allows the quickly and easily hADSCs and hOESCs isolation from human adipose tissue and nasal biopsies. The obtained cells can be cultured without altering their mesenchymal properties, suggesting the pluripotency nature of these cells and that they are a reliable source for regenerative medicine.
APA, Harvard, Vancouver, ISO, and other styles
10

Sunohara, Tadashi. "MicroRNA-based separation of cortico-fugal projection neuron-like cells derived from embryonic stem cells." Kyoto University, 2020. http://hdl.handle.net/2433/253176.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Books on the topic "Adipose-derived stem cells, regenerative medicine"

1

Illouz, Yves-Gerard, and Aris Sterodimas, eds. Adipose Stem Cells and Regenerative Medicine. Berlin, Heidelberg: Springer Berlin Heidelberg, 2011. http://dx.doi.org/10.1007/978-3-642-20012-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Illouz, Yves-Gérard. Adipose Stem Cells and Regenerative Medicine. Berlin, Heidelberg: Springer-Verlag Berlin Heidelberg, 2011.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
3

Illouz, Yves-Gerard, and Aris Sterodimas. Adipose Stem Cells and Regenerative Medicine. Springer, 2016.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
4

Li, Guo, Jinfeng Liao, and Yunfeng Lin. Adipose-Derived Stem Cells: Clinical Applications, Biological Characteristics and Therapeutic Potential in Regenerative Medicine. Nova Science Publishers, Incorporated, 2017.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
5

Allan, David S., and Dirk Strunk. Regenerative Therapy Using Blood-Derived Stem Cells. Humana Press, 2011.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
6

Allan, David S., and Dirk Strunk. Regenerative Therapy Using Blood-Derived Stem Cells. Humana, 2014.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
7

Allan, David S., and Dirk Strunk. Regenerative Therapy Using Blood-Derived Stem Cells. Humana Press, 2011.

Find full text
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "Adipose-derived stem cells, regenerative medicine"

1

Brown, Lora L. "Adipose-Derived Stromal Stem Cells." In Regenerative Medicine, 49–60. Cham: Springer International Publishing, 2022. http://dx.doi.org/10.1007/978-3-030-75517-1_5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Pojda, Zygmunt. "Adipose-Derived Stem Cells for Therapeutic Applications." In Regenerative Medicine, 77–89. London: Springer London, 2014. http://dx.doi.org/10.1007/978-1-4471-6542-2_9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Li, Jie, Elizabeth C. Martin, and Jeffrey M. Gimble. "Adipose-Derived Stem/Stromal Cells." In Regenerative Medicine - from Protocol to Patient, 363–87. Cham: Springer International Publishing, 2016. http://dx.doi.org/10.1007/978-3-319-27610-6_14.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Aronowitz, Joel A., Daniel Oheb, Nathan Cai, Asli Pekcan, Bridget Winterhalter, and Joseph Clayton. "Esthetic Surgery Applications for Adipose-Derived Stem Cells." In Regenerative Medicine, 265–71. Cham: Springer International Publishing, 2022. http://dx.doi.org/10.1007/978-3-030-75517-1_26.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Dechesne, Claude A., Didier F. Pisani, Sébastien Goudenege, and Christian Dani. "Adipose-Derived Stem Cells and Skeletal Muscle Repair." In Stem Cells & Regenerative Medicine, 77–87. Totowa, NJ: Humana Press, 2010. http://dx.doi.org/10.1007/978-1-60761-860-7_5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Khan, Aamer. "Adipose-Derived Stem Cells and Cellular Enhancement." In Regenerative Medicine in Aesthetic Treatments, 31–36. London: CRC Press, 2021. http://dx.doi.org/10.1201/9781003001478-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Schroeder, Josh E., Shaul Beyth, and Meir Liebergall. "Orthopedic Use of Adipose-Derived Stem Cells." In Adipose Stem Cells and Regenerative Medicine, 181–91. Berlin, Heidelberg: Springer Berlin Heidelberg, 2011. http://dx.doi.org/10.1007/978-3-642-20012-0_17.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Locke, Michelle B., and Vaughan J. Feisst. "Human Adipose-Derived Stem Cells (ASC): Their Efficacy in Clinical Applications." In Regenerative Medicine, 135–49. London: Springer London, 2014. http://dx.doi.org/10.1007/978-1-4471-6542-2_13.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Lin, Ching-Shwun, and Tom F. Lue. "Adipose-Derived Stem Cells: Characterization and Application in Urology." In Adipose Stem Cells and Regenerative Medicine, 193–207. Berlin, Heidelberg: Springer Berlin Heidelberg, 2011. http://dx.doi.org/10.1007/978-3-642-20012-0_18.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Indumathi, Somasundaram, Rashmi Mishra, Radhakrishnan Harikrishnan, and Marappagounder Dhanasekaran. "Subcutaneous Adipose Tissue-Derived Stem Cells: Advancement and Applications in Regenerative Medicine." In Regenerative Medicine, 91–112. London: Springer London, 2014. http://dx.doi.org/10.1007/978-1-4471-6542-2_10.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Conference papers on the topic "Adipose-derived stem cells, regenerative medicine"

1

Thirumala, Sreedhar, Jeffrey M. Gimble, and Ram V. Devireddy. "Apoptotic Response and Differentiation Ability of Adipose Derived Stem Cells (ASCs) Frozen/Thawed in the Presence of Polyvinylpyrrolidone (PVP)." In ASME 2009 Summer Bioengineering Conference. American Society of Mechanical Engineers, 2009. http://dx.doi.org/10.1115/sbc2009-206375.

Full text
Abstract:
Effective techniques for the cryopreservation of human Adipose Stem Cells (ASCs) could increase the usefulness of these cells in tissue engineering and regenerative medicine. The traditional method of using fetal calf serum (FCS) and a chemical cryoprotectant like dimethylsulfoxide (DMSO) during freezing storage restricts the direct use of ASCs in regenerative medicine and reconstruction surgery. The objective of this study was to investigate the apoptotic, necrotic and viability response of ASCs frozen/thawed in the presence of a high molecular weight polymer, Polyvinylpyrrolidone (PVP) and in the absence of FCS. Post-freeze/thaw studies were also conducted to evaluate the effect of PVP on the in vitro osteogenic and adipogenic differentiation of ASCs. The results were compared with those obtained using the most commonly used cryopreservation media of 10%DMSO + 10% Dulbecco’s Modified Eagle Media (DMEM) + 80%FCS.
APA, Harvard, Vancouver, ISO, and other styles
2

Kumar, Arun, and Binil Starly. "Modeling Human Mesenchymal Stem Cell Expansion in Vertical Wheel Bioreactors Using Lactate Production Rate in Regenerative Medicine Biomanufacturing." In ASME 2016 11th International Manufacturing Science and Engineering Conference. American Society of Mechanical Engineers, 2016. http://dx.doi.org/10.1115/msec2016-8787.

Full text
Abstract:
Stem cells are critical components of regenerative medicine therapy. However, the therapy will require millions to billions of therapeutic stem cells. To address the need, we have recently cultured stem cells in 3D microgels and used them as a vehicle for cell expansion within a low shear stress rotating wheel type bioreactor within a 500ml volumetric setting. This study specifically highlights the cell encapsulation in microbead process, harvesting and operation of microbeads within a dynamic bioreactor environment. We have specifically encapsulated stem cells (human adipose derived) into microbeads prepared from alginate hydrogels via an electrostatic jetting process. This study highlights the effect of fabrication process parameters on end-point biological quality measures such as stem cell count and viability. We were able to maintain a >80% viability during the 21 day static culture period. We have also measured the concentration of metabolites produced during the expansion, specifically lactate production measured during specific time points within culture inside the rotating wheel bioreactor Future work will need to address predicting yields in higher volume settings, efficiency of harvest and a more detailed description of the hydrodynamics affecting stem cell growth.
APA, Harvard, Vancouver, ISO, and other styles
3

Sudama, Hiroki, Atsushi Ogawa, Kei Saito, Wataru Ando, Norimasa Nakamura, and Hiromichi Fujie. "Effect of Shear Stress on Extracellular Matrix Production of Synovium-Derived Cells." In ASME 2009 Summer Bioengineering Conference. American Society of Mechanical Engineers, 2009. http://dx.doi.org/10.1115/sbc2009-206331.

Full text
Abstract:
It is well known that various fibrous tissue such as tendons and ligaments functionally adapt to dynamic and static loads. Although a variety of biomechanical studies have been done to deterimine the mechanism of remodeling in fibrous tissues, it was difficult to obtain detailed information because of complicated condstitution of the tissues. We have developed a stem cell-based self-assembled tissue (scSAT) [1] for tissue engineering. Since the scSAT is consisted of synovium-derived mesenchyaml stem cells and their native extracellular matrix, it is a good experimental model to determine the process of remodeling of fibrous tisues. However, the response of shear stress to the scSAT specimen has not been determined so far, although such data are important for understanding of soft tissue remodeling and for improvement of regenerative medicine. Therefore, the present study was performed to determine the effect of shear stress on the extracellular matrix production of synovium-derived cells including mesenchymal stem cells.
APA, Harvard, Vancouver, ISO, and other styles
4

Emura, Ryo, Atsushi Ogawa, Kei Saito, Wataru Ando, Norimasa Nakamura, and Hiromichi Fujie. "Effect of Compressive Load on the Mechanical Property of a Stem Cell Based Self-Assembled Tissue Derived From Synovium." In ASME 2009 Summer Bioengineering Conference. American Society of Mechanical Engineers, 2009. http://dx.doi.org/10.1115/sbc2009-205893.

Full text
Abstract:
Articular cartilage has superior functions such as impact absorption and low friction, although their healing capacities are limited. It is one of potential options for the repair of articular cartilage to use cell-based therapies. We have been developing a novel tissue-engineering technique for the repair of cartilage which involves a stem cell-based self-assembled tissue (scSAT) derived from synovium. As the scSAT is a scaffold-free contrust composed of cells with their native extracellular matrix, it is free from concern regarding long-term immunological effects. The scSAT is expressed as tissue engineered construct (TEC) when it is used for cartilage repair. Previous studies indicated that the mechanical properties of cartilage-like tissues repaired using the scSAT were slightly inferiorer to those of normal cartilage. We have a hypothesis that the mechanical properties of the cartilage-like tissues are improved if the scSAT is subjected to an adequate compressive stimulation in vitro before implantation. The present study was conducted as a preliminary study to determine whether static compression improves the mechanical property of the scSAT for more advanced regenerative medicine to cartilage injuries and degeneration.
APA, Harvard, Vancouver, ISO, and other styles
5

Bockman, Matthew D., Igor I. Katkov, Stephen B. Jones, Vsevolod Katkov, and Ilya Yakhnenko. "ComfortFreezer™: A Benchtop LN2–Free Programmable Freezer for Cryopreservation of Adherent Cells in Multi-Well Plates for Cell-Based High Content Screening." In ASME 2012 International Mechanical Engineering Congress and Exposition. American Society of Mechanical Engineers, 2012. http://dx.doi.org/10.1115/imece2012-85469.

Full text
Abstract:
Human pluripotent stem cells (hPSCs) and their progeny such as hPSC-derived cardiomyocytes and neural cells hold great potential as a source for cell therapy and regenerative medicine, as well can be effectively used for high high content screening (HCS) of drug candidates and for toxicity tests. Cryopreservation (CP), storage, and shipment of the cells are key elements for eventual clinical, pharmaceutical and environmental applications, which will require large numbers of quality controlled and ready for use cells. Traditionally, the cells are frozen in suspensions of either fully dissociated cells) or loosely associated clusters such as clumps of hPSCs, clusters of beaters”of cardiomyocytes, (“or neurospheres of neural precursors. Beside logistical inconvenience for some applications such as HCS, additional manipulation with the cells (detachment, dissociation and centrifugation) can introduce substantial stress to the cells prior to freezing and after thawing, which per se may tremendously decrease the cell cryosurvival and functionality. Here, we are presenting ComfortFreezer™, a novel bench-top device specifically designed for cryopreservation in multi-well plates for cell-based high content screening (HCS), which combines a liquid-nitrogen (LN2) free programmable freezer This cryogenic equipemnt can bring serious advantage for HCS in drug screening, environmental toxicity evaluation, and other variety of HCS-based applications.
APA, Harvard, Vancouver, ISO, and other styles
6

Alegre-Aguarón, Elena, Sonal R. Sampat, Perry J. Hampilos, J. Chloë Bulinski, James L. Cook, Lewis M. Brown, and Clark T. Hung. "Biomarker Identification Under Growth Factor Priming for Cartilage Tissue Engineering." In ASME 2012 Summer Bioengineering Conference. American Society of Mechanical Engineers, 2012. http://dx.doi.org/10.1115/sbc2012-80374.

Full text
Abstract:
Adult articular cartilage has a poor healing capacity, which has lead to intense research toward development of cell-based therapies for cartilage repair. The destruction of articular cartilage results in osteoarthritis (OA), which affects about 27 million Americans. In order to create functional tissue, it is essential to mimic the native environment by optimizing expansion protocols. Cell passaging and priming with chemical or physical factors are often necessary steps in cell-based strategies for regenerative medicine [1]. The ability to identify biomarkers that can act as predictors of cells with a high capacity to form functional engineered cartilage will permit optimization of protocols for cartilage tissue engineering using different cell sources. Recent investigations have shown that chondrocytes and synovium-derived stem cells (SDSCs) are promising cell sources for cartilage repair [2,3]. The analysis of gene expression and comparative proteomics, which defines the differences in expression of proteins among different biological states, provides a potentially powerful tool in this effort [4]. The aim of this study was to investigate the impact of growth factor priming in 2D canine chondrocytes and SDSCs cultures by identifying differentially regulated biomarkers, which can correlate to functional tissue elaboration in 3D.
APA, Harvard, Vancouver, ISO, and other styles
7

Tandon, N., B. Goh, A. Marsano, P. H. G. Chao, C. Montouri-Sorrentino, J. Gimble, and G. Vunjak-Novakovic. "Alignment and elongation of human adipose-derived stem cells in response to direct-current electrical stimulation." In 2009 Annual International Conference of the IEEE Engineering in Medicine and Biology Society. IEEE, 2009. http://dx.doi.org/10.1109/iembs.2009.5333142.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography