74 results on '"Biophysical stimulation"'
Search Results
2. Acceleration of experimental endochondral ossification by biophysical stimulation of the progenitor cell pool.
- Author
-
Aaron RK and Ciombor DM
- Subjects
- Animals, Bone Density physiology, Bone Matrix metabolism, Calcium analysis, Cartilage chemistry, Electric Stimulation, Electromagnetic Fields, Extracellular Matrix chemistry, Extracellular Matrix physiology, Glycosaminoglycans analysis, Male, Mesoderm physiology, Osteogenesis radiation effects, Periodicity, Rats, Stem Cells physiology, Cartilage cytology, Extracellular Matrix radiation effects, Osteogenesis physiology, Stem Cells radiation effects
- Abstract
Endochondral ossification can be modulated by a number of biochemical and biophysical stimuli. This study uses the experimental model of decalcified bone matrix-induced endochondral ossification to examine the effect of one biophysical stimulus, an electromagnetic field, on chondrogenesis, calcification, and osteogenesis. A temporal acceleration and quantitative increase in sulfate incorporation, glycosaminoglycan content, and calcification suggests that the stimulation of endochondral ossification is due to an increase in extracellular matrix synthesis. The locus of that stimulation is identified in the mesenchymal stage of endochondral bone development, and stimulation at this stage is essential for accelerated bone formation. The data suggest that enhanced differentiation of mesenchymal stem cells present at this stage is most likely responsible for the increase in extracellular matrix synthesis and bone maturation.
- Published
- 1996
- Full Text
- View/download PDF
3. Microenvironmental Regulation of Stem Cell Behavior Through Biochemical and Biophysical Stimulation
- Author
-
Bogyu, Choi, Deogil, Kim, Inbo, Han, and Soo-Hong, Lee
- Subjects
Stem Cells ,Humans ,Cell Differentiation ,Cell Lineage ,Stem Cell Niche ,Cellular Reprogramming - Abstract
Stem cells proliferate by undergoing self-renewal and differentiate into multiple cell lineages in response to biochemical and biophysical stimuli. Various biochemical cues such as growth factors, nucleic acids, chemical reagents, and small molecules have been used to induce stem cell differentiation or reprogramming or to maintain their pluripotency. Moreover, biophysical cues such as matrix stiffness, substrate topography, and external stress and strain play a major role in modulating stem cell behavior. In this chapter, we have summarized microenvironmental regulation of stem cell behavior through biochemical and biophysical stimulation.
- Published
- 2018
4. Mesenchymal stem cells as therapeutic target of biophysical stimulation for the treatment of musculoskeletal disorders.
- Author
-
Viganò, Marco, Sansone, Valerio, d'Agostino, Maria Cristina, Romeo, Pietro, Perucca Orfei, Carlotta, and de Girolamo, Laura
- Subjects
- *
TREATMENT effectiveness , *ELECTROMAGNETIC fields , *STEM cells , *ULTRASONIC therapy ,TREATMENT of musculoskeletal system diseases - Abstract
Background: Musculoskeletal disorders are regarded as a major cause of worldwide morbidity and disability, and they result in huge costs for national health care systems. Traditional therapies frequently turned out to be poorly effective in treating bone, cartilage, and tendon disorders or joint degeneration. As a consequence, the development of novel biological therapies that can treat more effectively these conditions should be the highest priority in regenerative medicine. Main body of the abstract: Mesenchymal stem cells (MSCs) represent one of the most promising tools in musculoskeletal tissue regenerative medicine, thanks to their proliferation and differentiation potential and their immunomodulatory and trophic ability. Indeed, MSC-based approaches have been proposed for the treatment of almost all orthopedic conditions, starting from different cell sources, alone or in combination with scaffolds and growth factors, and in one-step or two-step procedures. While all these approaches would require cell harvesting and transplantation, the possibility to stimulate the endogenous MSCs to enhance their tissue homeostasis activity represents a less-invasive and cost-effective therapeutic strategy. Nowadays, the role of tissue-specific resident stem cells as possible therapeutic target in degenerative pathologies is underinvestigated. Biophysical stimulations, and in particular extracorporeal shock waves treatment and pulsed electromagnetic fields, are able to induce proliferation and support differentiation of MSCs from different origins and affect their paracrine production of growth factors and cytokines. Short conclusions: The present review reports the attempts to exploit the resident stem cell potential in musculoskeletal pathologies, highlighting the role of MSCs as therapeutic target of currently applied biophysical treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
5. Pulsed Electromagnetic Fields in Bone Healing: Molecular Pathways and Clinical Applications.
- Author
-
Caliogna L, Medetti M, Bina V, Brancato AM, Castelli A, Jannelli E, Ivone A, Gastaldi G, Annunziata S, Mosconi M, and Pasta G
- Subjects
- Databases, Factual, Electromagnetic Fields, Humans, Osteogenesis genetics, Signal Transduction genetics, Stem Cells metabolism, Fractures, Bone radiotherapy, Magnetic Field Therapy methods, Osteogenesis radiation effects, Signal Transduction radiation effects, Stem Cells radiation effects
- Abstract
In this article, we provide an extensive review of the recent literature of the signaling pathways modulated by Pulsed Electromagnetic Fields (PEMFs) and PEMFs clinical application. A review of the literature was performed on two medical electronic databases (PubMed and Embase) from 3 to 5 March 2021. Three authors performed the evaluation of the studies and the data extraction. All studies for this review were selected following these inclusion criteria: studies written in English, studies available in full text and studies published in peer-reviewed journal. Molecular biology, identifying cell membrane receptors and pathways involved in bone healing, and studying PEMFs target of action are giving a solid basis for clinical applications of PEMFs. However, further biology studies and clinical trials with clear and standardized parameters (intensity, frequency, dose, duration, type of coil) are required to clarify the precise dose-response relationship and to understand the real applications in clinical practice of PEMFs.
- Published
- 2021
- Full Text
- View/download PDF
6. Acceleration of experimental endochondral ossification by biophysical stimulation of the progenitor cell pool
- Author
-
Roy K. Aaron and Deborah K. Mc Ciombor
- Subjects
Male ,Periodicity ,Bone Matrix ,Extracellular matrix ,Mesoderm ,Electromagnetic Fields ,Bone Density ,Osteogenesis ,medicine ,Animals ,Orthopedics and Sports Medicine ,Endochondral ossification ,Glycosaminoglycans ,Chemistry ,Cartilage ,Stem Cells ,Mesenchymal stem cell ,Anatomy ,Chondrogenesis ,medicine.disease ,Electric Stimulation ,Cell biology ,Extracellular Matrix ,Rats ,medicine.anatomical_structure ,Intramembranous ossification ,Bone maturation ,Calcium ,Calcification - Abstract
Endochondral ossification can be modulated by a number of biochemical and biophysical stimuli. This study uses the experimental model of decalcified bone matrix-induced endochondral ossification to examine the effect of one biophysical stimulus, an electromagnetic field, on chondrogenesis, calcification, and osteogenesis. A temporal acceleration and quantitative increase in sulfate incorporation, glycosaminoglycan content, and calcification suggests that the stimulation of endochondral ossification is due to an increase in extracellular matrix synthesis. The locus of that stimulation is identified in the mesenchymal stage of endochondral bone development, and stimulation at this stage is essential for accelerated bone formation. The data suggest that enhanced differentiation of mesenchymal stem cells present at this stage is most likely responsible for the increase in extracellular matrix synthesis and bone maturation.
- Published
- 1996
7. Pulsed Electromagnetic Fields in Bone Healing: Molecular Pathways and Clinical Applications
- Author
-
Valentina Bina, Salvatore Annunziata, Alessandro Ivone, Giulia Gastaldi, Eugenio Jannelli, Gianluigi Pasta, Alice Maria Brancato, Laura Caliogna, Alberto Castelli, Marta Medetti, Mario Mosconi, Caliogna, L., Medetti, M., Bina, V., Brancato, A. M., Castelli, A., Jannelli, E., Ivone, A., Gastaldi, G., Annunziata, S., Mosconi, M., and Pasta, G.
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Databases, Factual ,QH301-705.5 ,Magnetic Field Therapy ,osteogenic differentiation ,Bone healing ,Review ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,Fractures, Bone ,0302 clinical medicine ,Electromagnetic Fields ,bone regeneration ,Osteogenesis ,medicine ,Humans ,Medical physics ,Physical and Theoretical Chemistry ,Biology (General) ,Bone regeneration ,Molecular Biology ,QD1-999 ,Spectroscopy ,pulsed electromagnetic fields (PEMFs) ,biophysical stimulation ,Osteogenesi ,Stem Cells ,Organic Chemistry ,General Medicine ,fracture repair ,fracture healing ,Computer Science Applications ,Clinical trial ,Clinical Practice ,Chemistry ,030104 developmental biology ,Electromagnetic Field ,030220 oncology & carcinogenesis ,Human ,Signal Transduction - Abstract
In this article, we provide an extensive review of the recent literature of the signaling pathways modulated by Pulsed Electromagnetic Fields (PEMFs) and PEMFs clinical application. A review of the literature was performed on two medical electronic databases (PubMed and Embase) from 3 to 5 March 2021. Three authors performed the evaluation of the studies and the data extraction. All studies for this review were selected following these inclusion criteria: studies written in English, studies available in full text and studies published in peer-reviewed journal. Molecular biology, identifying cell membrane receptors and pathways involved in bone healing, and studying PEMFs target of action are giving a solid basis for clinical applications of PEMFs. However, further biology studies and clinical trials with clear and standardized parameters (intensity, frequency, dose, duration, type of coil) are required to clarify the precise dose-response relationship and to understand the real applications in clinical practice of PEMFs.
- Published
- 2021
8. Nanosecond pulsed electric fields enhance mesenchymal stem cells differentiation via DNMT1-regulated OCT4/NANOG gene expression.
- Author
-
Li, Kejia, Ning, Tong, Wang, Hao, Jiang, Yangzi, Zhang, Jue, and Ge, Zigang
- Subjects
- *
MESENCHYMAL stem cell differentiation , *MESENCHYMAL stem cells , *ELECTRIC fields , *GENE expression , *DNA methyltransferases , *STEM cells - Abstract
Background: Multiple strategies have been proposed to promote the differentiation potential of mesenchymal stem cells (MSCs), which is the fundamental property in tissue formation and regeneration. However, these strategies are relatively inefficient that limit the application. In this study, we reported a novel and efficient strategy, nanosecond pulsed electric fields (nsPEFs) stimulation, which can enhance the trilineage differentiation potential of MSCs, and further explained the mechanism behind. Methods: We used histological staining to screen out the nsPEFs parameters that promoted the trilineage differentiation potential of MSCs, and further proved the effect of nsPEFs by detecting the functional genes. In order to explore the corresponding mechanism, we examined the expression of pluripotency genes and the methylation status of their promoters. Finally, we targeted the DNA methyltransferase which was affected by nsPEFs. Results: The trilineage differentiation of bone marrow-derived MSCs was significantly enhanced in vitro by simply pre-treating with 5 pulses of nsPEFs stimulation (energy levels as 10 ns, 20 kV/cm; 100 ns, 10 kV/cm), due to that the nsPEFs demethylated the promoters of stem cell pluripotency genes OCT4 and NANOG through instantaneous downregulation of DNA methylation transferase 1 (DNMT1), thereby increasing the expression of OCT4 and NANOG for up to 3 days, and created a treatment window period of stem cells. Conclusions: In summary, nsPEFs can enhance MSCs differentiation via the epigenetic regulation and could be a safe and effective strategy for future stem cell application. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
9. Advanced maturation by electrical stimulation: Differences in response between C2C12 and primary muscle progenitor cells
- Author
-
Mark J. Post, Kang Yuen Rosaria-Chak, Frank P. T. Baaijens, K.J.M. Boonen, Marloes L. P. Langelaan, Daisy W. J. van der Schaft, Soft Tissue Biomech. & Tissue Eng., Fysiologie, and RS: CARIM School for Cardiovascular Diseases
- Subjects
cell line vs. primary cell source ,sarcomere assembly ,Biomedical Engineering ,Medicine (miscellaneous) ,Stimulation ,Biology ,muscle progenitor cells ,Sarcomere ,Polymerase Chain Reaction ,Cell Line ,Biomaterials ,Mice ,Myosin ,Myocyte ,Animals ,Progenitor cell ,skeletal muscle tissue engineering ,biophysical stimulation ,Matrigel ,Myosin Heavy Chains ,maturation ,Muscles ,Stem Cells ,Immunohistochemistry ,Electric Stimulation ,Cell biology ,Stem cell ,C2C12 ,Biomedical engineering - Abstract
Skeletal muscle tissue engineering still does not result in the desired functional properties and texture as preferred for regenerative medicine and meat production applications. Electrical stimulation has been appropriately used as a tool to advance muscle cell maturation in vitro, thereby simulating nerve stimulation, as part of the muscle cell niche in vivo. We first investigated the effects of electrical stimulation protocols in two-dimensional (2D) monolayers of C2C12 and translated these protocols to a three-dimensional (3D) model system, based on a collagen type I/Matrigel(?) hydrogel. More importantly, we addressed the ongoing debate of the translation of results found in cell lines (C2C12) to a primary cell source [muscle progenitor cells (MPCs)] in our 3D system. Striking differences in maturation level were found between the different cell sources. Constructs with MPCs were much more mature than C2C12 constructs, based on developed cross-striations and expression levels of mature myosin heavy chain (MHC) isoforms. Overall, electrical stimulation, when optimally timed, accelerated sarcomere assembly in both 2D and 3D. In addition, MPC constructs were more susceptible to the electrical stimulus, resulting in a shift of MHC expression to slower isoforms.
- Published
- 2010
10. Designing magnetic microcapsules for cultivation and differentiation of stem cell spheroids.
- Author
-
Gwon, Kihak, Dharmesh, Ether, Nguyen, Kianna M., Schornack, Anna Marie R., de Hoyos-Vega, Jose M., Ceylan, Hakan, Stybayeva, Gulnaz, Peterson, Quinn P., and Revzin, Alexander
- Subjects
PLURIPOTENT stem cells ,HUMAN stem cells ,IRON oxide nanoparticles ,STEM cells ,CELL differentiation ,TISSUE engineering - Abstract
Human pluripotent stem cells (hPSCs) represent an excellent cell source for regenerative medicine and tissue engineering applications. However, there remains a need for robust and scalable differentiation of stem cells into functional adult tissues. In this paper, we sought to address this challenge by developing magnetic microcapsules carrying hPSC spheroids. A co-axial flow-focusing microfluidic device was employed to encapsulate stem cells in core-shell microcapsules that also contained iron oxide magnetic nanoparticles (MNPs). These microcapsules exhibited excellent response to an external magnetic field and could be held at a specific location. As a demonstration of utility, magnetic microcapsules were used for differentiating hPSC spheroids as suspension cultures in a stirred bioreactor. Compared to standard suspension cultures, magnetic microcapsules allowed for more efficient media change and produced improved differentiation outcomes. In the future, magnetic microcapsules may enable better and more scalable differentiation of hPSCs into adult cell types and may offer benefits for cell transplantation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Effects of BMP9 and pulsed electromagnetic fields on the proliferation and osteogenic differentiation of human periodontal ligament stem cells.
- Author
-
Wang T, Wang P, Cao Z, Wang X, Wang D, Shen Y, Jing D, Luo E, and Tang W
- Subjects
- Adolescent, Cell Proliferation drug effects, Cell Proliferation radiation effects, Child, Extracellular Matrix metabolism, Humans, Minerals metabolism, Stem Cells cytology, Up-Regulation drug effects, Electromagnetic Fields, Growth Differentiation Factor 2 pharmacology, Osteogenesis drug effects, Periodontal Ligament cytology, Stem Cells drug effects, Stem Cells radiation effects
- Abstract
Periodontal ligament stem cells (PDLSCs) have been confirmed to have self-renewal capacity and multidifferentiation potential and are good candidates for periodontal tissue regeneration. Pulsed electromagnetic field (PEMF) has been demonstrated to promote osteogenesis in non-union fractures, partly by regulating mesenchymal stem cells or osteoblast activity. However, there is no report about the osteo-inductive effect of PEMF stimulation on human PDLSCs (hPDLSCs). Thus, we tested the hypothesis that PEMF biophysical stimulation alone has an influence on the proliferation and osteogenic differentiation of hPDLSCs. To detect the osteo-inductive potential of bone morphogenetic protein (BMP9), we transfected the STRO-1
+ /CD146+ hPDLCSs with BMP9-expressing recombinant adenoviruses. We examined the proliferation and osteogenic differentiation of hPDLSCs treated with either PEMF (15 Hz, 1 h daily, different intensities), or BMP9, or both stimuli. Cell counting kit-8 (CCK-8) assay showed that PEMF of different intensities had no effect on the proliferation of hPDLSCs and did not enhance the proliferative capability of BMP9-transfected cells. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting showed that the combination of both PEMFs (1.8 or 2.4 mT) and BMP9 stimulation had a synergistic effect on early and intermediate osteogenic genes and protein expressions of runt-related transcription factor 2, alkaline phosphatase, osteopontin, and late mineralized extracellular matrix formation in hPDLSCs. Bioelectromagnetics. 38:63-77, 2017. © 2016 Wiley Periodicals, Inc., (© 2016 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
12. Effects of the pulsed electromagnetic field PST® on human tendon stem cells: a controlled laboratory study.
- Author
-
Randelli P, Menon A, Ragone V, Creo P, Alfieri Montrasio U, Perucca Orfei C, Banfi G, Cabitza P, Tettamanti G, and Anastasia L
- Subjects
- Cell Physiological Phenomena radiation effects, Cells, Cultured, Humans, Magnetic Field Therapy, Stem Cells cytology, Stem Cells radiation effects, Tendons cytology
- Abstract
Background: Current clinical procedures for rotator cuff tears need to be improved, as a high rate of failure is still observed. Therefore, new approaches have been attempted to stimulate self-regeneration, including biophysical stimulation modalities, such as low-frequency pulsed electromagnetic fields, which are alternative and non-invasive methods that seem to produce satisfying therapeutic effects. While little is known about their mechanism of action, it has been speculated that they may act on resident stem cells. Thus, the purpose of this study was to evaluate the effects of a pulsed electromagnetic field (PST®) on human tendon stem cells (hTSCs) in order to elucidate the possible mechanism of the observed therapeutic effects., Methods: hTSCs from the rotator cuff were isolated from tendon biopsies and cultured in vitro. Then, cells were exposed to a 1-h PST® treatment and compared to control untreated cells in terms of cell morphology, proliferation, viability, migration, and stem cell marker expression., Results: Exposure of hTSCs to PST® did not cause any significant changes in proliferation, viability, migration, and morphology. Instead, while stem cell marker expression significantly decreased in control cells during cell culturing, PST®-treated cells did not have a significant reduction of the same markers., Conclusions: While PST® did not have significant effects on hTSCs proliferation, the treatment had beneficial effects on stem cell marker expression, as treated cells maintained a higher expression of these markers during culturing. These results support the notion that PST® treatment may increase the patient stem cell regenerative potential.
- Published
- 2016
- Full Text
- View/download PDF
13. Recent Progress in Engineering Mesenchymal Stem Cell Differentiation.
- Author
-
Halim, Alexander, Ariyanti, Agnes Dwi, Luo, Qing, and Song, Guanbin
- Subjects
- *
MESENCHYMAL stem cell differentiation , *STEM cells , *CELL differentiation , *CELLULAR therapy , *MESENCHYMAL stem cells - Abstract
Due to the ability to differentiate into variety of cell types, mesenchymal stem cells (MSCs) hold promise as source in cell-based therapy for treating injured tissue and degenerative diseases. The potential use of MSCs to replace or repair damaged tissues may depend on the efficient differentiation protocols to derive specialized cells without any negative side effects. Identification of appropriate cues that support the lineage-specific differentiation of stem cells is critical for tissue healing and cellular therapy. Recently, a number of stimuli have been utilized to direct the differentiation of stem cells. Biochemical stimuli such as small molecule, growth factor and miRNA have been traditionally used to regulate the fate of stem cells. In recent years, many studies have reported that biophysical stimuli including cyclic mechanical strain, fluid shear stress, microgravity, electrical stimulation, matrix stiffness and topography can also be sensed by stem cells through mechanical receptors, thus affecting the stem cell behaviors including their differentiation potential. In this paper, we review all the most recent literature on the application of biochemical and biophysical cues on regulating MSC differentiation. An extensive literature search was done using electronic database (Medline/Pubmed). Although there are still some challenges that need to be taken into consideration before translating these methods into clinics, biochemical and biophysical stimulation appears to be an attractive method to manipulate the lineage commitment of MSCs. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
14. Biophysical cues enhance myogenesis of human adipose derived stem/stromal cells.
- Author
-
Yilgor Huri P, Cook CA, Hutton DL, Goh BC, Gimble JM, DiGirolamo DJ, and Grayson WL
- Subjects
- Adipocytes physiology, Cell Differentiation, Cell Proliferation, Cells, Cultured, Humans, Physical Stimulation methods, Stress, Mechanical, Tissue Engineering methods, Adipocytes cytology, Mechanotransduction, Cellular physiology, Muscle Development physiology, Muscle Fibers, Skeletal cytology, Muscle Fibers, Skeletal physiology, Stem Cells cytology, Stem Cells physiology
- Abstract
Adipose-derived stem/stromal cell (ASC)-based tissue engineered muscle grafts could provide an effective alternative therapy to autografts - which are limited by their availability - for the regeneration of damaged muscle. However, the current myogenic potential of ASCs is limited by their low differentiation efficiency into myoblasts. The aim of this study was to enhance the myogenic response of human ASCs to biochemical cues by providing biophysical stimuli (11% cyclic uniaxial strain, 0.5 Hz, 1h/day) to mimic the cues present in the native muscle microenvironment. ASCs elongated and fused upon induction with myogenic induction medium alone. Yet, their myogenic characteristics were significantly enhanced with the addition of biophysical stimulation; the nuclei per cell increased approximately 4.5-fold by day 21 in dynamic compared to static conditions (23.3 ± 7.3 vs. 5.2 ± 1.6, respectively), they aligned at almost 45° to the direction of strain, and exhibited significantly higher expression of myogenic proteins (desmin, myoD and myosin heavy chain). These results demonstrate that mimicking the biophysical cues inherent to the native muscle microenvironment in monolayer ASC cultures significantly improves their differentiation along the myogenic lineage., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
15. Adipose tissue as a stem cell source for musculoskeletal regeneration.
- Author
-
Gimble JM, Grayson W, Guilak F, Lopez MJ, and Vunjak-Novakovic G
- Subjects
- Cell Separation methods, Dinoprostone metabolism, Genetic Therapy methods, Humans, Stem Cells cytology, Stromal Cells cytology, Stromal Cells physiology, Adipose Tissue cytology, Cell Differentiation physiology, Musculoskeletal Development physiology, Regeneration physiology, Stem Cell Transplantation methods, Stem Cells physiology, Tissue Engineering methods
- Abstract
Adipose tissue is an abundant, easily accessible, and reproducible cell source for musculo-skeletal regenerative medicine applications. Initial derivation steps yield a heterogeneous population of cells of stromal vascular fraction (SVF) cells. Subsequent adherent selection of the SVF results in a relatively homogeneous population of adipose-derived stromal/stem cells (ASCs) capable of adipogenic, chondrogenic, myogenic, and osteogenic differentiation in vitro on scaffolds in bioreactors and in vivo in pre-clinical animal models. Unlike hematopoietic cells, ASCs do not elicit a robust lymphocyte reaction and instead release immunosuppressive factors, such as prostaglandin E2. These immunomodulatory features suggest that allogeneic and autologous ASCs will engraft successfully for tissue regeneration purposes. The differentiation and expansion potential of ASCs can be modified by growth factors, bio-inductive scaffolds, and bioreactors providing environmental control and biophysical stimulation. Gene therapy approaches using lentiviral transduction can be used to direct differentiation of ASCs to particular lineages. We discuss the utility of ASCs for musculo-skeletal tissue repair and some of the technologies that can be implemented to unlock the full regenerative potential of these highly valuable cells.
- Published
- 2011
- Full Text
- View/download PDF
16. Considerations for enhanced mesenchymal stromal/stem cell myogenic commitment in vitro.
- Author
-
Grobbelaar, Simone, Mercier, Anne E., van den Bout, Iman, Durandt, Chrisna, and Pepper, Michael S.
- Subjects
MYOBLASTS ,STEM cells ,CELL determination ,PROGENITOR cells ,CELL culture ,REGENERATIVE medicine ,CARTILAGE regeneration - Abstract
The generation of tissue from stem cells is an alluring concept as it holds a number of potential applications in clinical therapeutics and regenerative medicine. Mesenchymal stromal/stem cells (MSCs) can be isolated from a number of different somatic sources, and have the capacity to differentiate into adipogenic, osteogenic, chondrogenic, and myogenic lineages. Although the first three have been extensively investigated, there remains a paucity of literature on the latter. This review looks at the various strategies available in vitro to enhance harvested MSC commitment and differentiation into the myogenic pathway. These include chemical inducers, myogenic‐enhancing cell culture substrates, and mechanical and dynamic culturing conditions. Drawing on information from embryonic and postnatal myogenesis from somites, satellite, and myogenic progenitor cells, the mechanisms behind the chemical and mechanical induction strategies can be studied, and the sequential gene and signaling cascades can be used to monitor the progression of myogenic differentiation in the laboratory. Increased understanding of the stimuli and signaling mechanisms in the initial stages of MSC myogenic commitment will provide tools with which we can enhance their differentiation efficacy and advance the process to clinical translation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Rapid Magneto-Sonoporation of Adipose-Derived Cells.
- Author
-
Filippi, Miriam, Dasen, Boris, and Scherberich, Arnaud
- Subjects
MAGNETIC resonance imaging ,IRON oxide nanoparticles ,CELL differentiation ,ULTRASONIC imaging ,STEM cells - Abstract
By permeabilizing the cell membrane with ultrasound and facilitating the uptake of iron oxide nanoparticles, the magneto-sonoporation (MSP) technique can be used to instantaneously label transplantable cells (like stem cells) to be visualized via magnetic resonance imaging in vivo. However, the effects of MSP on cells are still largely unexplored. Here, we applied MSP to the widely applicable adipose-derived stem cells (ASCs) for the first time and investigated its effects on the biology of those cells. Upon optimization, MSP allowed us to achieve a consistent nanoparticle uptake (in the range of 10 pg/cell) and a complete membrane resealing in few minutes. Surprisingly, this treatment altered the metabolic activity of cells and induced their differentiation towards an osteoblastic profile, as demonstrated by an increased expression of osteogenic genes and morphological changes. Histological evidence of osteogenic tissue development was collected also in 3D hydrogel constructs. These results point to a novel role of MSP in remote biophysical stimulation of cells with focus application in bone tissue repair. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
18. The Nuclear Option: Evidence Implicating the Cell Nucleus in Mechanotransduction.
- Author
-
Szczesny, Spencer E. and Mauck, Robert L.
- Subjects
- *
MECHANOTRANSDUCTION (Cytology) , *CELL nuclei , *CELL membranes - Abstract
Biophysical stimuli presented to cells via microenvironmental properties (e.g., alignment and stiffness) or external forces have a significant impact on cell function and behavior. Recently, the cell nucleus has been identified as a mechanosensitive organelle that contributes to the perception and response to mechanical stimuli. However, the specific mechanotransduction mechanisms that mediate these effects have not been clearly established. Here, we offer a comprehensive review of the evidence supporting (and refuting) three hypothetical nuclear mechanotransduction mechanisms: physical reorganization of chromatin, signaling at the nuclear envelope, and altered cytoskeletal structure/tension due to nuclear remodeling. Our goal is to provide a reference detailing the progress that has been made and the areas that still require investigation regarding the role of nuclear mechanotransduction in cell biology. Additionally, we will briefly discuss the role that mathematical models of cell mechanics can play in testing these hypotheses and in elucidating how biophysical stimulation of the nucleus drives changes in cell behavior. While force-induced alterations in signaling pathways involving lamina-associated polypeptides (LAPs) (e.g., emerin and histone deacetylase 3 (HDAC3)) and transcription factors (TFs) located at the nuclear envelope currently appear to be the most clearly supported mechanism of nuclear mechanotransduction, additional work is required to examine this process in detail and to more fully test alternative mechanisms. The combination of sophisticated experimental techniques and advanced mathematical models is necessary to enhance our understanding of the role of the nucleus in the mechanotransduction processes driving numerous critical cell functions. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
19. Magnetoresponsive stem cell spheroid-based cartilage recovery platform utilizing electromagnetic fields.
- Author
-
Yoo, Ami, Go, Gwangjun, Nguyen, Kim Tien, Lee, Kyungmin, Min, Hyun-Ki, Kang, Byungjeon, Kim, Chang-Sei, Han, Jiwon, Park, Jong-Oh, and Choi, Eunpyo
- Subjects
- *
ELECTROMAGNETIC fields , *CARTILAGE , *STEM cells , *MAGNETIC control , *CARTILAGE cells , *CARTILAGE regeneration - Abstract
• We propose magnetoresponsive stem cell spheroid (MR-SCS)-based cartilage recovery platform for targeting and biophysical stimulation. • Locomotion of MR-SCS that was mediated by the electromagnetic actuation system was successfully demonstrated in 3D phantom and ex vivo models • Low-frequency electromagnetic field stimulation of MR-SCS resulted in increased expression levels of cartilage specific markers. • Ex vivo evaluation revealed an improvement in the regeneration of cartilage tissue in Low-frequency electromagnetic field stimulation. Mesenchymal stem cells (MSCs) provide a promising source for cartilage tissue regeneration strategies. The use of MSCs for such strategies, however, remains challenging due to the low targeting and low chondrogenic differentiation efficiency of these cells to the desired site. In an attempt to overcome such problems, we propose the use of a magnetoresponsive stem cell spheroid (MR-SCS)-based cartilage recovery platform that allows for precise targeting using an electromagnetic actuation (EMA) system to provide magnetic control and low-frequency electromagnetic field (LF-EMF) to allow for biophysical stimulation to promote chondrogenic differentiation. MR-SCSs were fabricated from mouse-derived MSCs that were labeled with magnetic nanoparticles (MNPs) using 3D culture methods, and these particles exhibited no cytotoxicity and did not affect chondrogenic differentiation. Locomotion of MR-SCS that was mediated by the EMA system was successfully demonstrated in 3D phantom and ex vivo models. Additionally, LF-EMF stimulation of MR-SCS resulted in increased expression levels of cartilage specific markers, collagen type II, SOX9, and Aggrecan. Finally, histological evaluation revealed an apparent improvement in the regeneration of cartilage tissue in an ex vivo model of the porcine femur in response to LF-EMF stimulation. These results suggest that our experimental platform consisting of MR-SCSs that are subjected to EMA and LF-EMF stimulation may provide a promising therapeutic system for cartilage tissue regeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
20. Biophysical Modulation of Mesenchymal Stem Cell Differentiation in the Context of Skeletal Repair.
- Author
-
Hung, Clark T., Racine-Avila, Jennifer, Pellicore, Matthew J., and Aaron, Roy
- Subjects
- *
MESENCHYMAL stem cell differentiation , *STRAINS & stresses (Mechanics) , *EXTRACELLULAR matrix , *CELL differentiation , *SHEARING force - Abstract
A prominent feature of the skeleton is its ability to remodel in response to biophysical stimuli and to repair under varied biophysical conditions. This allows the skeleton considerable adaptation to meet its physiological roles of stability and movement. Skeletal cells and their mesenchymal precursors exist in a native environment rich with biophysical signals, and they sense and respond to those signals to meet organismal demands of the skeleton. While mechanical strain is the most recognized of the skeletal biophysical stimuli, signaling phenomena also include fluid flow, hydrostatic pressure, shear stress, and ion-movement-related electrokinetic phenomena including, prominently, streaming potentials. Because of the complex interactions of these electromechanical signals, it is difficult to isolate the significance of each. The application of external electrical and electromagnetic fields allows an exploration of the effects of these stimuli on cell differentiation and extra-cellular matrix formation in the absence of mechanical strain. This review takes a distinctly translational approach to mechanistic and preclinical studies of differentiation and skeletal lineage commitment of mesenchymal cells under biophysical stimulation. In vitro studies facilitate the examination of isolated cellular responses while in vivo studies permit the observation of cell differentiation and extracellular matrix synthesis. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
21. Neurogenesis-on-Chip: Electric field modulated transdifferentiation of human mesenchymal stem cell and mouse muscle precursor cell coculture.
- Author
-
Naskar, Sharmistha, Kumaran, Viswanathan, Markandeya, Yogananda S., Mehta, Bhupesh, and Basu, Bikramjit
- Subjects
- *
HUMAN stem cells , *MESENCHYMAL stem cells , *ELECTRIC fields , *MUSCLE cells , *STEM cells , *MYOBLASTS - Abstract
A number of bioengineering strategies, using biophysical stimulation, are being explored to guide the human mesenchymal stem cells (hMScs) into different lineages. In this context, we have limited understanding on the transdifferentiation of matured cells to another functional-cell type, when grown with stem cells, in a constrained cellular microenvironment under biophysical stimulation. While addressing such aspects, the present work reports the influence of the electric field (EF) stimulation on the phenotypic and functionality modulation of the coculture of murine myoblasts (C2C12) with hMScs [hMSc:C2C12=1:10] in a custom designed polymethylmethacrylate (PMMA) based microfluidic device with in-built metal electrodes. The quantitative and qualitative analysis of the immunofluorescence study confirms that the cocultured cells in the conditioned medium with astrocytic feed, exhibit differentiation towards neural-committed cells under biophysical stimulation in the range of the endogenous physiological electric field strength (8 ± 0.06 mV/mm). The control experiments using similar culture protocols revealed that while C2C12 monoculture exhibited myotube-like fused structures, the hMScs exhibited the neurosphere-like clusters with SOX2, nestin, βIII-tubulin expression. The electrophysiological study indicates the significant role of intercellular calcium signalling among the differentiated cells towards transdifferentiation. Furthermore, the depolarization induced calcium influx strongly supports neural-like behaviour for the electric field stimulated cells in coculture. The intriguing results are explained in terms of the paracrine signalling among the transdifferentiated cells in the electric field stimulated cellular microenvironment. In summary, the present study establishes the potential for neurogenesis on-chip for the coculture of hMSc and C2C12 cells under tailored electric field stimulation, in vitro. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. ETV2 promotes osteogenic differentiation of human dental pulp stem cells through the ERK/MAPK and PI3K-Akt signaling pathways.
- Author
-
Li, Jing, Du, Haoran, Ji, Xin, Chen, Yihan, Li, Yishuai, Heng, Boon Chin, and Xu, Jianguang
- Subjects
DENTAL pulp ,TRANSCRIPTION factors ,STEM cells ,CELLULAR signal transduction ,MITOGEN-activated protein kinases ,OSTEOBLASTS ,BONE regeneration ,GENE expression - Abstract
Background: The repair of cranio-maxillofacial bone defects remains a formidable clinical challenge. The Ets variant 2 (ETV2) transcription factor, which belongs to the E26 transformation-specific (ETS) family, has been reported to play a key role in neovascularization. However, the role of ETV2 in the osteogenesis of human dental pulp stem cells (hDPSCs) remains unexplored. Methods: Transgenic overexpression of ETV2 was achieved using a lentiviral vector, based on a Dox-inducible system. The effects of Dox-induced overexpression of ETV2 on the osteogenesis of hDPSCs were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR), western blot, immunofluorescence staining, alkaline phosphatase (ALP) staining, and Alizarin Red S (ARS) staining. Additionally, RNA-sequencing (RNA-Seq) analysis was performed to analyze the underlying mechanisms of ETV2-induced osteogenesis. Additionally, the role of ETV2 overexpression in bone formation in vivo was validated by animal studies with a rat calvarial defect model and a nude mice model. Results: Our results demonstrated that ETV2 overexpression significantly upregulated the mRNA and protein expression levels of osteogenic markers, markedly enhanced ALP activity, and promoted matrix mineralization of hDPSCs. Moreover, the results of RNA-Seq analysis and western blot showed that the ERK/MAPK and PI3K-Akt signaling pathways were activated upon transgenic overexpression of ETV2. The enhanced osteogenic differentiation of hDPSCs due to ETV2 overexpression was partially reversed by treatment with inhibitors of ERK/MAPK or PI3K-AKT signaling. Furthermore, the results of in vivo studies demonstrated that ETV2 overexpression improved bone healing in a rat calvarial defect model and increased ectopic bone formation in nude mice. Conclusions: Collectively, our results indicated that ETV2 overexpression exerted positive effects on the osteogenesis of hDPSCs, at least partially via the ERK/MAPK and PI3K/AKT signaling pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
23. Cell therapy efficacy and safety in treating tendon disorders: a systemic review of clinical studies.
- Author
-
Mirghaderi, Seyed Peyman, Valizadeh, Zahra, Shadman, Kimia, Lafosse, Thibault, Oryadi-Zanjani, Leila, Yekaninejad, Mir Saeed, and Nabian, Mohammad Hossein
- Subjects
CELLULAR therapy ,JUMPER'S knee ,ROTATOR cuff ,ACHILLES tendinitis ,TENDONS ,MESENCHYMAL stem cells ,PAIN management - Abstract
Purpose: Despite substantial animal evidence, cell therapy in humans remains in its infancy. The purpose of this study was to examine the potential therapeutic effects and safety of cell therapy in the treatment of tendon disorders. Methods: According to the PRISMA guideline, a systematic review was performed on clinical studies concerning cell therapy in tendon disorders. A comprehensive search including the 5 databases of MEDLINE, Embase, Scopus, Web of Science, and Cochrane Library until December 2021 was carried out and associated with hand searching. The quality of the eligible studies was assessed using the tools suggested by Cochrane recommendations. Qualitative synthesis was performed in 2 tables and discussed separately for rotator cuff, elbow, patella, Achilles, and gluteal tendons. Results: Through 6017 records, 22 studies were included in the qualitative synthesis, including 658 patients. All the studies administered autologous cells, except one that used allogenic adipose-derived mesenchymal stem cells (Allogenic AD-MSC). Almost all studies demonstrated the safety of cell injection in their follow-up period with no serious side effects or immunologic reactions, with only a few related minor adverse events in some cases. The included studies showed the effectiveness of cell injection in tendinopathies of different sites, rotator cuff, elbow, patella, Achilles, and gluteal tendons. Among the rotator cuff studies, 4 comparative studies claimed that cell therapy is a more efficient treatment with a lower retear rate and pain level compared to the control group. However, one study found no differences between the groups. No controlled study has been performed on elbow tendinopathies, but 5 case series demonstrated the effectiveness of cell injection in elbow tendon disorders. For Achilles tendinopathies, only one randomized controlled trial (RCT) found that both cell therapy and control groups showed significant pain reduction and functional improvement with no statistical differences at the 6 months follow-up, but the cell therapy group had improved faster at earlier follow-ups. Patellar tendinopathy was studied in 2 RCTs, one did not show a significant difference and the other showed superior improvement compared to controls. Conclusion: Cell therapy showed promising results and the available evidence suggests that it is safe at several sites of tendon disease. Based on available evidence, cell therapy should be suggested in specific conditions at each site. To approve cell therapy for tendon diseases, randomized clinical trials are required with a large sample size and long-term follow-ups. Level of evidence: IV [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
24. Adult stem cell sources for skeletal and smooth muscle tissue engineering.
- Author
-
Salemi, Souzan, Prange, Jenny A., Baumgartner, Valentin, Mohr-Haralampieva, Deana, and Eberli, Daniel
- Subjects
SMOOTH muscle ,STEM cells ,STEM cell transplantation ,SKELETAL muscle ,ADULTS ,CELL physiology - Abstract
Introduction: Tissue engineering is an innovative field with enormous developments in recent years. These advances are not only in the understanding of how stem cells can be isolated, cultured and manipulated but also in their potential for clinical applications. Thus, tissue engineering when applied to skeletal and smooth muscle cells is an area that bears high benefit for patients with muscular diseases or damage. Most of the recent research has been focused on use of adult stem cells. These cells have the ability to rejuvenate and repair damaged tissues and can be derived from different organs and tissue sources. Recently there are several different types of adult stem cells, which have the potential to function as a cell source for tissue engineering of skeletal and smooth muscles. However, to build neo‐tissues there are several challenges which have to be addressed, such as the selection of the most suitable stem cell type, isolation techniques, gaining control over its differentiation and proliferation process. Conclusion: The usage of adult stem cells for muscle engineering applications is promising. Here, we summarize the status of research on the use of adult stem cells for cell transplantation in experimental animals and humans. In particular, the application of skeletal and smooth muscle engineering in pre-clinical and clinical trials will be discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. Differentiation of adipose-derived stem cells to chondrocytes using electrospraying.
- Author
-
Nosoudi, Nasim, Hart, Christoph, McKnight, Ian, Esmaeilpour, Mehdi, Ghomian, Taher, Zadeh, Amir, Raines, Regan, and Ramirez Vick, Jaime E.
- Subjects
TRANSFORMING growth factors ,BONE morphogenetic proteins ,STEM cells ,CARTILAGE regeneration ,MULTIPOTENT stem cells ,CARTILAGE cells - Abstract
An important challenge in the fabrication of tissue engineered constructs for regenerative medical applications is the development of processes capable of delivering cells and biomaterials to specific locations in a consistent manner. Electrospraying live cells has been introduced in recent years as a cell seeding method, but its effect on phenotype nor genotype has not been explored. A promising candidate for the cellular component of these constructs are human adipose-derived stem cells (hASCs), which are multipotent stem cells that can be differentiated into fat, bone, and cartilage cells. They can be easily and safely obtained from adipose tissue, regardless of the age and sex of the donor. Moreover, these cells can be maintained and expanded in culture for long periods of time without losing their differentiation capacity. In this study, hASCs directly incorporated into a polymer solution were electrosprayed, inducing differentiation into chondrocytes, without the addition of any exogenous factors. Multiple studies have demonstrated the effects of exposing hASCs to biomolecules—such as soluble growth factors, chemokines, and morphogens—to induce chondrogenesis. Transforming growth factors (e.g., TGF-β) and bone morphogenetic proteins are particularly known to play essential roles in the induction of chondrogenesis. Although growth factors have great therapeutic potential for cell-based cartilage regeneration, these growth factor-based therapies have presented several clinical complications, including high dose requirements, low half-life, protein instability, higher costs, and adverse effects in vivo. The present data suggests that electrospraying has great potential as hASCs-based therapy for cartilage regeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
26. Polarized M2 macrophages induced by mechanical stretching modulate bone regeneration of the craniofacial suture for midfacial hypoplasia treatment.
- Author
-
Liang, Wei, Ding, Pengbing, Qian, Jiaying, Li, Guan, Lu, Enhang, and Zhao, Zhenmin
- Subjects
MACROPHAGES ,SUTURES ,SUTURING ,ANIMAL disease models ,STEM cells ,STRETCH (Physiology) ,BONE regeneration ,FRACTURE healing - Abstract
The underlying mechanism of the trans-sutural distraction osteogenesis (TSDO) technique as an effective treatment that improves the symptoms of midfacial hypoplasia syndromes is not clearly understood. Increasing findings in the orthopedics field indicate that macrophages are mechanically sensitive and their phenotypes can respond to mechanical cues. However, how macrophages respond to mechanical stretching and consequently influence osteoblast differentiation of suture-derived stem cells (SuSCs) remains unclear, particularly during the TSDO process. In the present study, we established a TSDO rat model to determine whether and how macrophages were polarized in response to stretching and consequently affected bone regeneration of the suture frontal edge. Notably, after performing immunofluorescence, RNA-sequencing, and micro-computed tomography, it was demonstrated that macrophages are first recruited by various chemokines factors and polarized to the M2 phenotype upon optimal stretching. The latter in turn regulates SuSC activity and facilitates bone regeneration in sutures. Moreover, when the activated M2 macrophages were suppressed by pharmacological manipulation, new bone microarchitecture could rarely be detected under mechanical stretching and the expansion of the sutures was clear. Additionally, macrophages achieved M2 polarization in response to the optimal mechanical stretching (10%, 0.5 Hz) and strongly facilitated SuSC osteogenic differentiation and human umbilical vein endothelial cell angiogenesis using an indirect co-culture system in vitro. Collectively, this study revealed the mechanical stimulation-immune response-bone regeneration axis and clarified at least in part how sutures achieve bone regeneration in response to mechanical force. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
27. MiR-596 inhibits osteoblastic differentiation and cell proliferation by targeting Smad3 in steroid-induced osteonecrosis of femoral head.
- Author
-
Fu, Ligong, Liu, Huawei, and Lei, Weijun
- Subjects
CELL proliferation ,BONE marrow ,OSTEONECROSIS ,CARRIER proteins ,CELL differentiation ,GENE expression ,GLUCOCORTICOIDS ,POLYMERASE chain reaction ,STEM cells ,STEROIDS ,WESTERN immunoblotting ,DEXAMETHASONE ,FEMUR head ,OSTEOBLASTS ,MICRORNA ,PHARMACODYNAMICS - Abstract
Background: It is reported that miR-596 has a potential diagnostic value for non-traumatic osteonecrosis of the femoral head (NOFH), but its underlying mechanisms in NOFH is unclear. Methods: The expression of miR-596 and Smad3 was detected by western blot and quantitative real-time PCR. The relationship between the two molecules was explored using Dual-Luciferase Reporter Assay. Glucocorticoid (GC)—dexamethasone, was used to induce bone marrow mesenchymal stem cell (BMSC) osteogenic differentiation, and the effects of miR-596 on BMSC osteogenic differentiation and proliferation were determined. Results: MiR-596 expression was upregulated, while Smad3 expression was inhibited in the bone marrow samples of patients with steroid-induced osteonecrosis of femoral head (SANFH). Overexpression of miR-596 inhibited the proliferation and osteogenic differentiation of BMSCs induced by GC. Meanwhile, the opposite results were observed in the miR-596 inhibitor group. In addition, Smad3 was a target gene of miR-596, and negatively regulated by miR-596. The promotion effect of the miR-596 inhibitor on BMSC proliferation and osteogenic differentiation was reversed by si-Smad3. Conclusion: MiR-596 can suppress GC-BMSC osteoblastic differentiation and proliferation by regulating Smad3 expression. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
28. New Insights on Mechanical Stimulation of Mesenchymal Stem Cells for Cartilage Regeneration.
- Author
-
Ravalli, Silvia, Szychlinska, Marta Anna, Lauretta, Giovanni, and Musumeci, Giuseppe
- Subjects
CARTILAGE regeneration ,CARTILAGE cells ,MESENCHYMAL stem cells ,STEM cell culture ,BIOLOGICAL systems ,HYDROSTATIC pressure ,STEM cells - Abstract
Successful tissue regeneration therapies require further understanding of the environment in which the cells are destined to be set. The aim is to structure approaches that aspire to a holistic view of biological systems and to scientific reliability. Mesenchymal stem cells represent a valuable resource for cartilage tissue engineering, due to their chondrogenic differentiation capacity. Promoting chondrogenesis, not only by growth factors but also by exogenous enhancers such as biomechanics, represents a technical enhancement. Tribological evaluation of the articular joint has demonstrated how mechanical stimuli play a pivotal role in cartilage repair and participate in the homeostasis of this tissue. Loading stresses, physiologically experienced by chondrocytes, can upregulate the production of proteins like glycosaminoglycan or collagen, fundamental for articular wellness, as well as promote and preserve cell viability. Therefore, there is a rising interest in the development of bioreactor devices that impose compression, shear stress, and hydrostatic pressure on stem cells. This strategy aims to mimic chondrogenesis and overcome complications like hypertrophic phenotyping and inappropriate mechanical features. This review will analyze the dynamics inside the joint, the natural stimuli experienced by the chondrocytes, and how the biomechanical stimuli can be applied to a stem cell culture in order to induce chondrogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
29. Cell-Based Bone Reconstruction Therapies--Principles of Clinical Approaches.
- Author
-
Meyer, Ulrich, Wiesmann, Hans-Peter, Berr, Karin, Kübler, Norbert R., and Handschel, Jörg
- Subjects
BONE remodeling ,BONE regeneration ,STEM cell research ,MAXILLOFACIAL surgery ,AUTOTRANSPLANTATION ,CLINICAL trials - Abstract
Cell-based bone tissue engineering is a rapidly evolving therapy option in bone reconstruction strategies. Some cell-driven approaches, especially the bio physical stimulation of the host cell population surrounded by the bone defect, are common treatment methods in maxillo facial surgery. Others, such as autologous cell implantation, have now gained acceptance for clinical trials. More advanced or complex therapeutical options (extracorporeal tissue engineering, stem cell use, genetic engineering) have been tested in preclinical investigations but have not reached the level of clinical use. Two different aspects are of special relevance in cell-based bone reconstruction therapies. The source of cells used to regenerate bone (discussed in detail in a complementary review in this issue of The International Journal of Oral and Maxillofacial Implants) as well as the principal approach of a cell-driven bone regeneration therapy influence the outcome of such engineering strategies. All of the cell-driven repair strategies are under intensive investigation in an effort to provide surgeons with a limitless supply of tissue for bone repair and reconstruction in future procedures. An overview of the basic biological aspects as well as the inherent constraints of different cell-based approaches are given in this paper. [ABSTRACT FROM AUTHOR]
- Published
- 2006
30. Human adipose-derived Mesenchymal stem cells, low-intensity pulsed ultrasound, or their combination for the treatment of knee osteoarthritis: study protocol for a first-in-man randomized controlled trial.
- Author
-
Nasb, Mohammad, Liangjiang, Huang, Gong, Chenzi, and Hong, Chen
- Subjects
MESENCHYMAL stem cells ,RANDOMIZED controlled trials ,CLINICAL trial registries ,STEM cell treatment ,KNEE ,INTRA-articular injections - Abstract
Background: Human adipose-derived Mesenchymal stem cells (HADMSCs) have proven their efficacy in treating osteoarthritis (OA), in earlier preclinical and clinical studies. As the tissue repairers are under the control of mechanical and biochemical signals, improving regeneration outcomes using such signals has of late been the focus of attention. Among mechanical stimuli, low-intensity pulsed ultrasound (LIPUS) has recently shown promise both in vitro and in vivo. This study will investigate the potential of LIPUS in enhancing the regeneration process of an osteoarthritic knee joint.Methods: This study involves a prospective, randomized, placebo-controlled, and single-blind trial based on the SPIRIT guidelines, and aims to recruit 96 patients initially diagnosed with knee osteoarthritis, following American College of Rheumatology criteria. Patients will be randomized in a 1:1:1 ratio to receive Intraarticular HADMSCs injection with LIPUS, Intraarticular HADMSCs injection with shame LIPUS, or Normal saline with LIPUS. The primary outcome is Western Ontario and McMaster Universities Index of OA (WOMAC) score, while the secondary outcomes will be other knee structural changes, and lower limb muscle strength such as the knee cartilage thickness measured by MRI. Blinded assessments will be performed at baseline (1 month prior to treatment), 1 month, 3 months, and 6 months following the interventions.Discussion: This trial will be the first clinical study to comprehensively investigate the safety and efficacy of LIPUS on stem cell therapy in OA patients. The results may provide evidence of the effectiveness of LIPUS in improving stem cell therapy and deliver valuable information for the design of subsequent trials.Trial Registration: This study had been prospectively registered with the Chinese Clinical Trials Registry. registration number: ChiCTR1900025907 at September 14, 2019. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
31. Tendon and Ligament Healing and Current Approaches to Tendon and Ligament Regeneration.
- Author
-
Leong, Natalie L., Kator, Jamie L., Clemens, Thomas L., James, Aaron, Enamoto‐Iwamoto, Motomi, and Jiang, Jie
- Subjects
TENDON injury healing ,LIGAMENTS ,TENDONS ,LIGAMENT injuries ,TENDON injuries ,STEM cells - Abstract
Ligament and tendon injuries are common problems in orthopedics. There is a need for treatments that can expedite nonoperative healing or improve the efficacy of surgical repair or reconstruction of ligaments and tendons. Successful biologically‐based attempts at repair and reconstruction would require a thorough understanding of normal tendon and ligament healing. The inflammatory, proliferative, and remodeling phases, and the cells involved in tendon and ligament healing will be reviewed. Then, current research efforts focusing on biologically‐based treatments of ligament and tendon injuries will be summarized, with a focus on stem cells endogenous to tendons and ligaments. Statement of clinical significance: This paper details mechanisms of ligament and tendon healing, as well as attempts to apply stem cells to ligament and tendon healing. Understanding of these topics could lead to more efficacious therapies to treat ligament and tendon injuries. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:7–12, 2020 [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
32. Insights into the Molecular Mechanisms Regulating Cell Behavior in Response to Magnetic Materials and Magnetic Stimulation in Stem Cell (Neurogenic) Differentiation.
- Author
-
Mocanu-Dobranici, Alexandra-Elena, Costache, Marieta, and Dinescu, Sorina
- Subjects
MAGNETIC materials ,NUCLEAR membranes ,CALCIUM channels ,STEM cells ,CELLULAR evolution ,CYCLIC adenylic acid ,MAGNETIC nanoparticles - Abstract
Magnetic materials and magnetic stimulation have gained increasing attention in tissue engineering (TE), particularly for bone and nervous tissue reconstruction. Magnetism is utilized to modulate the cell response to environmental factors and lineage specifications, which involve complex mechanisms of action. Magnetic fields and nanoparticles (MNPs) may trigger focal adhesion changes, which are further translated into the reorganization of the cytoskeleton architecture and have an impact on nuclear morphology and positioning through the activation of mechanotransduction pathways. Mechanical stress induced by magnetic stimuli translates into an elongation of cytoskeleton fibers, the activation of linker in the nucleoskeleton and cytoskeleton (LINC) complex, and nuclear envelope deformation, and finally leads to the mechanical regulation of chromatin conformational changes. As such, the internalization of MNPs with further magnetic stimulation promotes the evolution of stem cells and neurogenic differentiation, triggering significant changes in global gene expression that are mediated by histone deacetylases (e.g., HDAC 5/11), and the upregulation of noncoding RNAs (e.g., miR-106b~25). Additionally, exposure to a magnetic environment had a positive influence on neurodifferentiation through the modulation of calcium channels' activity and cyclic AMP response element-binding protein (CREB) phosphorylation. This review presents an updated and integrated perspective on the molecular mechanisms that govern the cellular response to magnetic cues, with a special focus on neurogenic differentiation and the possible utility of nervous TE, as well as the limitations of using magnetism for these applications. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
33. 干细胞心肌向分化成熟过程中的物理刺激方法.
- Author
-
张泽茜, 伍佳琪, 樊瑜波, and 郑丽沙
- Abstract
Copyright of Chinese Journal of Tissue Engineering Research / Zhongguo Zuzhi Gongcheng Yanjiu is the property of Chinese Journal of Tissue Engineering Research and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2019
- Full Text
- View/download PDF
34. A comprehensive overview on utilizing electromagnetic fields in bone regenerative medicine.
- Author
-
Azadian, Esmaeel, Arjmand, Bahar, Khodaii, Zohreh, and Ardeshirylajimi, Abdolreza
- Subjects
TISSUE engineering ,MESENCHYMAL stem cells ,REGENERATIVE medicine ,CELL proliferation ,ELECTROMAGNETIC fields - Abstract
Stem cells are one of the most important sources to develope a new strategy for repairing bone lesions through tissue engineering. Osteogenic differentiation of stem cells can be affected by various factors such as biological, chemical, physiological, and physical ones. The application of ELF-EMFs has been the subject of many research in bone tissue engineering and evidence suggests that this exogenous physical stimulus can promote osteogenic differentiation in several types of cells. The purpose of this paper is to review the current knowledge on the effects of EMFs on stem cells in bone tissue engineering studies. We recapitulated and analyzed 39 articles that were focused on the application of EMFs for bone tissue engineering purposes. We tabulated scattered information from these articles for easy use and tried to provide an overview of conducted research and identify the knowledge gaps in the field. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
35. Flexure-Based Device for Cyclic Strain-Mediated Osteogenic Differentiation
- Author
-
Kyung Shin Kang, Woon-Jae Yong, Young Hun Jeong, Jong-Won Rhie, Dong-Woo Cho, and Jung Min Hong
- Subjects
Calcium Phosphates ,Polyesters ,Biomedical Engineering ,Biocompatible Materials ,Cell Separation ,Extracellular matrix ,chemistry.chemical_compound ,Polylactic Acid-Polyglycolic Acid Copolymer ,Osteogenesis ,Physiology (medical) ,Animals ,Humans ,Nanotechnology ,Lactic Acid ,Osteopontin ,Bone regeneration ,Tissue Scaffolds ,biology ,Chemistry ,business.industry ,Stem Cells ,Mesenchymal stem cell ,Cell Differentiation ,Structural engineering ,In vitro ,Rats ,PLGA ,Adipose Tissue ,biology.protein ,Alkaline phosphatase ,Stress, Mechanical ,Stem cell ,business ,Polyglycolic Acid ,Biomedical engineering - Abstract
Biophysical strain has been applied widely for bone regeneration. However, application of low-magnitude strains to cells on small-thickness scaffolds is problematic, especially in rodent calvarial defect models, because general translation systems have limitations in terms of generating low-magnitude smooth signals. To overcome these limitations, we developed an in vitro biophysical-stimulation platform for stimulation of cells on small-thickness scaffolds for rodent calvarial bone defects. The customized flexure-based translational nanoactuator enables generation of low-magnitude smooth signals at the subnano- to micrometer-scale. This nanoactuator, which is equipped with a piezoelectric actuator, is suitable for biological applications because it can generate friction-free motion with a high resolution. Moreover, its operation without wear or deterioration eliminates contamination factors in cell culture environments. The developed in vitro biophysical-stimulation platform using these nanoactuators showed predictable operational characteristics. Also, a few-micrometer sinusoidal signal was generated successfully without any distortion. Three-dimensional scaffolds fitting the critical-size rat calvarial defect model were fabricated using poly(caprolactone), poly(lactic-co-glycolic acid), and tricalcium phosphate. Runt-related transcription factor 2 expression was increased upon stimulation of human adipose-derived stem cells (ASCs) on these scaffolds were stimulated in the in vitro biophysical-stimulation platform. Additionally, the use of this platform resulted in up-regulation of alkaline phosphate, osteopontin, and osterix expression compared to the non-stimulated group. These preliminary in vitro results suggest that the biophysical environment provided by the in vitro biophysical-stimulation platform influences the osteogenic differentiation of ASCs.
- Published
- 2013
- Full Text
- View/download PDF
36. The Effect of Scaffold Modulus on the Morphology and Remodeling of Fetal Mesenchymal Stem Cells.
- Author
-
Rufaihah, Abdul Jalil, Cheyyatraivendran, Suganya, Mazlan, Muhammad Danial Mohd, Lim, Kenrich, Chong, Mark Seow Khoon, Mattar, Citra Nurfarah Zaini, Chan, Jerry Kok Yen, Kofidis, Theodoros, and Seliktar, Dror
- Subjects
STEM cells ,MESENCHYMAL stem cells ,HYDROGELS in medicine ,INTEGRINS ,PHYSIOLOGY - Abstract
Hydrogel materials have been successfully used as matrices to explore the role of biophysical and biochemical stimuli in directing stem cell behavior. Here, we present our findings on the role of modulus in guiding bone marrow fetal mesenchymal stem cell (BMfMSC) fate determination using semi-synthetic hydrogels made from PEG-fibrinogen (PF). The BMfMSCs were cultivated in the PF for up to 2 weeks to study the influence of matrix modulus (i.e., cross-linking density of the PF) on BMfMSC survival, morphology and integrin expression. Both two-dimensional (2D) and three-dimensional (3D) culture conditions were employed to examine the BMfMSCs as single cells or as cell spheroids. The hydrogel modulus affected the rate of BMfMSC metabolic activity, the integrin expression levels and the cell morphology, both as single cells and as spheroids. The cell seeding density was also found to be an important parameter of the system in that high densities were favorable in facilitating more cell-to-cell contacts that favored higher metabolic activity. Our findings provide important insight about design of a hydrogel scaffold that can be used to optimize the biological response of BMfMSCs for various tissue engineering applications. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
37. Concise Review on Clinical Applications of Conditioned Medium Derived from Human Umbilical Cord-Mesenchymal Stem Cells (UC-MSCs).
- Author
-
Sriramulu, Sushmitha, Banerjee, Antara, Di Liddo, Rosa, Jothimani, Ganesan, Gopinath, Madhumala, Murugesan, Ramachandran, Marotta, Francesco, and Pathak, Surajit
- Subjects
MESENCHYMAL stem cells ,MULTIPOTENT stem cells ,REGENERATIVE medicine - Abstract
In recent years, mesenchymal stem cells have provoked much attentiveness in the field of regenerative medicine because of their differentiation potential and the capability to facilitate tissue repair via the emancipation of biologically active molecules. They have gained interest because of their distinctive curative properties. Mesenchymal stem cells are isolated from the Wharton's jelly part of umbilical cord possessing higher proliferation capacity, immunomodulatory activity, plasticity, as well as self-renewal capacity than the mesenchymal stem cells from various origins, and it is considered to be the best resource for allogeneic transplantation. The isolated umbilical cord-derived mesenchymal stem cells are cultured in the Dulbecco's Modified Eagle's Medium, and thereby it begins to release soluble factors into the medium during the period of culture which is termed as conditioned medium. This conditioned media has both differentiation capacity and therapeutic functions. Thus, it can be able to differentiate the cells into different lineages and the paracrine effect of these cells helps in replacement of the damaged cells. This medium may accord to optimization of diagnostic and prognostic systems as well as the generation of novel and targeted therapeutic perspectives. [ABSTRACT FROM AUTHOR]
- Published
- 2018
38. The effect of pulsed electromagnetic field exposure on osteoinduction of human mesenchymal stem cells cultured on nano-TiO2 surfaces.
- Author
-
Bloise, Nora, Petecchia, Loredana, Ceccarelli, Gabriele, Fassina, Lorenzo, Usai, Cesare, Bertoglio, Federico, Balli, Martina, Vassalli, Massimo, Cusella De Angelis, Maria Gabriella, Gavazzo, Paola, Imbriani, Marcello, and Visai, Livia
- Subjects
OSTEOINDUCTION ,MESENCHYMAL stem cells ,ELECTROMAGNETIC fields ,TITANIUM oxides ,CELL physiology - Abstract
Human bone marrow-derived mesenchymal stem cells (hBM-MSCs) are considered a great promise in the repair and regeneration of bone. Considerable efforts have been oriented towards uncovering the best strategy to promote stem cells osteogenic differentiation. In previous studies, hBM-MSCs exposed to physical stimuli such as pulsed electromagnetic fields (PEMFs) or directly seeded on nanostructured titanium surfaces (TiO
2 ) were shown to improve their differentiation to osteoblasts in osteogenic condition. In the present study, the effect of a daily PEMF-exposure on osteogenic differentiation of hBM-MSCs seeded onto nanostructured TiO2 (with clusters under 100 nm of dimension) was investigated. TiO2 -seeded cells were exposed to PEMF (magnetic field intensity: 2 mT; intensity of induced electric field: 5 mV; frequency: 75 Hz) and examined in terms of cell physiology modifications and osteogenic differentiation. Results showed that PEMF exposure affected TiO2 -seeded cells osteogenesis by interfering with selective calcium-related osteogenic pathways, and greatly enhanced hBM-MSCs osteogenic features such as the expression of early/late osteogenic genes and protein production (e.g., ALP, COL-I, osteocalcin and osteopontin) and ALP activity. Finally, PEMF-treated cells resulted to secrete into conditioned media higher amounts of BMP-2, DCN and COL-I than untreated cell cultures. These findings confirm once more the osteoinductive potential of PEMF, suggesting that its combination with TiO2 nanostructured surface might be a great option in bone tissue engineering applications. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
39. Direct Control of Stem Cell Behavior Using Biomaterials and Genetic Factors.
- Author
-
Yoon, Jeong-Kee, Kang, Mi-Lan, Park, Joo Hyun, Lee, Kyoung-Mi, Shin, Young Min, Lee, Jin Woo, Kim, Hyun Ok, and Sung, Hak-Joon
- Subjects
BIOMATERIALS ,STEM cells ,CELLULAR therapy ,GENE transfection ,GRAFT versus host disease - Abstract
Stem cells have recently emerged as an important candidate for cell therapy. However, some major limitations still exist such as a small quantity of cell supply, senescence, and insufficient differentiation efficiency. Therefore, there is an unmet need to control stem cell behavior for better clinical performance. Since native microenvironment factors including stem cell niche, genetic factors, and growth factors direct stem cell fate cooperatively, user-specified
in vitro settings are required to understand the regulatory roles and effects of each factor, thereby applying the factors for improved cell therapy. Among others, various types of biomaterials and transfection method have been employed as key tools for development of thein vitro settings. This review focuses on the current strategies to improve stemness maintenance, direct differentiation, and reprogramming using biomaterials and genetic factors without any aids from additional biochemicals and growth factors. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
40. Progress of Regenerative Therapy in Orthopedics.
- Author
-
Pearlin, Nayak, Sunita, Manivasagam, Geetha, and Sen, Dwaipayan
- Abstract
Purpose of Review: To conduct a thorough appraisal of recent and inventive advances in the field of bone tissue engineering using biomaterials, cell-based research, along with the incorporation of biomimetic properties using surface modification of scaffolds.Recent Findings: This paper will provide an overview on different biomaterials and emerging techniques involved in the fabrication of scaffolds, brief description of signaling pathways involved in osteogenesis, and the effect of surface modification on the fate of progenitor cells.Summary: The current strategies used for regenerative medicine like cell therapy, gene transfer, and tissue engineering have opened numerous therapeutic avenues for the treatment of various disabling orthopedic disorders. Precise strategy utilized for the reconstruction, restoration, or repair of the bone-related tissues exploits cells, biomaterials, morphogenetic signals, and appropriate mechanical environment to provide the basic constituents required for creating new tissue. Combining all the above strategies in clinical trials would pave the way for successful “bench to bedside” transformation in bone healing. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
41. Regulating osteogenesis and adipogenesis in adipose-derived stem cells by controlling underlying substrate stiffness.
- Author
-
Zhang, Tao, Lin, Shiyu, Shao, Xiaoru, Shi, Sirong, Zhang, Qi, Xue, Changyue, Lin, Yunfeng, Zhu, Bofeng, and Cai, Xiaoxiao
- Subjects
ADIPOGENESIS ,TISSUE engineering ,STEM cells ,CELL proliferation ,POLYDIMETHYLSILOXANE - Abstract
Cells reside in a complex microenvironment (niche) in which the biochemical and biophysical properties of the extracellular matrix profoundly affect cell behavior. Extracellular stiffness, one important bio-mechanical characteristic of the cell niche, is important in regulating cell proliferation, migration, and lineage specification. However, the mechanism by which mechanical signals guide osteogenic and adipogenic commitment of stem cells remains difficult to dissect. To explore this question, we generated a range of polydimethylsiloxane-based matrices with differing degrees of stiffness that mimicked the stiffness seen in natural tissues and examined adipose stem cell morphology, spreading, vinculin expression, and differentiation along the osteogenic and adipogenic pathways. Rigid matrices allowed broader cell spreading, faster growth rate and stronger expression of vinculin in adipose-derived stem cells. In the presence of inductive culture media, stiffness-dependent osteogenesis and adipogenesis of the adipose stem cells indicated that there was a combinatorial effect of biophysical and biochemical cues; no such lineage specification was observed in normal media. Osteogenic differentiation behavior showed a correlation with matrix rigidity, as well as with elevated expression of RhoA, ROCK-1/-2, and related proteins in the Wnt/β-catenin pathway. The result provides a comprehensive understanding of how stem cells respond to the surrounding microenvironment and points to the fact that matrix stiffness is a critical element in biomaterial design and this will be an important advance in stem cell-based tissue engineering. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
42. A Poroelastic Model of a Fibrous-Porous Tissue Engineering Scaffold.
- Author
-
Yuan, Daniel, Somers, Sarah M., Grayson, Warren L., and Spector, Alexander A.
- Subjects
POROELASTICITY ,TISSUE engineering ,STEM cells ,EXTRACELLULAR matrix ,FATE mapping (Genetics) - Abstract
issue engineering scaffolds are used in conjunction with stem cells for the treatment of various diseases. A number of factors provided by the scaffolds affect the differentiation of stem cells. Mechanical cues that are part of the natural cellular microenvironment can both accelerate the differentiation toward particular cell lineages or induce differentiation to an alternative cell fate. Among such factors, there are externally applied strains and mechanical (stiffness and relaxation time) properties of the extracellular matrix. Here, the mechanics of a fibrous-porous scaffold is studied by applying a coordinated modeling and experimental approach. A force relaxation experiment is used, and a poroelastic model associates the relaxation process with the fluid diffusion through the fibrous matrix. The model parameters, including the stiffness moduli in the directions along and across the fibers as well as fluid diffusion time, are estimated by fitting the experimental data. The time course of the applied force is then predicted for different rates of loading and scaffold porosities. The proposed approach can help in a reduction of the technological and experimental efforts to produce 3-D scaffolds for regenerative medicine as well as in a higher accuracy of the estimation of the local factors sensed by stem cells. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
43. Basic science of osteoarthritis.
- Author
-
Cucchiarini, Magali, de Girolamo, Laura, Filardo, Giuseppe, Oliveira, J. Miguel, Orth, Patrick, Pape, Dietrich, and Reboul, Pascal
- Subjects
OSTEOARTHRITIS ,ARTICULAR cartilage ,MESENCHYMAL stem cells ,TISSUE engineering ,JOINT diseases ,EXTRACELLULAR matrix - Abstract
Osteoarthritis (OA) is a prevalent, disabling disorder of the joints that affects a large population worldwide and for which there is no definitive cure. This review provides critical insights into the basic knowledge on OA that may lead to innovative end efficient new therapeutic regimens. While degradation of the articular cartilage is the hallmark of OA, with altered interactions between chondrocytes and compounds of the extracellular matrix, the subchondral bone has been also described as a key component of the disease, involving specific pathomechanisms controlling its initiation and progression. The identification of such events (and thus of possible targets for therapy) has been made possible by the availability of a number of animal models that aim at reproducing the human pathology, in particular large models of high tibial osteotomy (HTO). From a therapeutic point of view, mesenchymal stem cells (MSCs) represent a promising option for the treatment of OA and may be used concomitantly with functional substitutes integrating scaffolds and drugs/growth factors in tissue engineering setups. Altogether, these advances in the fundamental and experimental knowledge on OA may allow for the generation of improved, adapted therapeutic regimens to treat human OA. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
44. Stem cells in articular cartilage regeneration.
- Author
-
Filardo, Giuseppe, Perdisa, Francesco, Roffi, Alice, Marcacci, Maurilio, and Kon, Elizaveta
- Subjects
ARTICULAR cartilage ,COMPARATIVE studies ,MEDLINE ,ONLINE information services ,REGENERATION (Biology) ,RESEARCH funding ,STEM cells ,SYSTEMATIC reviews - Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising option to treat articular defects and early osteoarthritis (OA) stages. However, both their potential and limitations for a clinical use remain controversial. Thus, the aim of this systematic review was to examine MSCs treatment strategies in clinical settings, in order to summarize the current evidence of their efficacy for the treatment of cartilage lesions and OA. Among the 60 selected studies, 7 were randomized, 13 comparative, 31 case series, and 9 case reports; 26 studies reported the results after injective administration, whereas 33 used surgical implantation. One study compared the two different modalities. With regard to the cell source, 20 studies concerned BMSCs, 17 ADSCs, 16 BMC, 5 PBSCs, 1 SDSCs, and 1 compared BMC versus PBSCs. Overall, despite the increasing literature on this topic, the evidence is still limited, in particular for high-level studies. On the other hand, the available studies allow to draw some indications. First, no major adverse events related to the treatment or to the cell harvest have been reported. Second, a clinical benefit of using MSCs therapies has been reported in most of the studies, regardless of cell source, indication, or administration method. This effectiveness has been reflected by clinical improvements and also positive MRI and macroscopic findings, whereas histologic features gave more controversial results among different studies. Third, young age, lower BMI, smaller lesion size for focal lesions, and earlier stages of OA joints have been shown to correlate with better outcomes, even though the available data strength does not allow to define clear cutoff values. Finally, definite trends can be observed with regard to the delivery method: currently cultured cells are mostly being administered by i.a. injection, while one-step surgical implantation is preferred for cell concentrates. In conclusion, while promising results have been shown, the potential of these treatments should be confirmed by reliable clinical data through double-blind, controlled, prospective and multicenter studies with longer follow-up, and specific studies should be designed to identify the best cell sources, manipulation, and delivery techniques, as well as pathology and disease phase indications. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
45. Rapid Magneto-Sonoporation of Adipose-Derived Cells
- Author
-
Arnaud Scherberich, Boris Dasen, and Miriam Filippi
- Subjects
cell labelling ,magnetic nanoparticles ,Technology ,Cell ,Adipose tissue ,ultrasounds ,Bone tissue ,osteogenesis ,Cell membrane ,chemistry.chemical_compound ,superparamagnetic iron oxide particles ,In vivo ,stem cells ,parasitic diseases ,medicine ,General Materials Science ,magneto-sonoporation ,stem cell ,bone tissue engineering ,Microscopy ,QC120-168.85 ,Communication ,QH201-278.5 ,Engineering (General). Civil engineering (General) ,Cell biology ,TK1-9971 ,medicine.anatomical_structure ,chemistry ,Descriptive and experimental mechanics ,Electrical engineering. Electronics. Nuclear engineering ,Stem cell ,TA1-2040 ,Sonoporation ,Iron oxide nanoparticles - Abstract
By permeabilizing the cell membrane with ultrasound and facilitating the uptake of iron oxide nanoparticles, the magneto-sonoporation (MSP) technique can be used to instantaneously label transplantable cells (like stem cells) to be visualized via magnetic resonance imaging in vivo. However, the effects of MSP on cells are still largely unexplored. Here, we applied MSP to the widely applicable adipose-derived stem cells (ASCs) for the first time and investigated its effects on the biology of those cells. Upon optimization, MSP allowed us to achieve a consistent nanoparticle uptake (in the range of 10 pg/cell) and a complete membrane resealing in few minutes. Surprisingly, this treatment altered the metabolic activity of cells and induced their differentiation towards an osteoblastic profile, as demonstrated by an increased expression of osteogenic genes and morphological changes. Histological evidence of osteogenic tissue development was collected also in 3D hydrogel constructs. These results point to a novel role of MSP in remote biophysical stimulation of cells with focus application in bone tissue repair., Materials, 14 (17), ISSN:1996-1944
- Published
- 2021
46. Corroboration of computational models for mechanoregulated stem cell differentiation.
- Author
-
Khayyeri, Hanifeh, Isaksson, Hanna, and Prendergast, Patrick J.
- Subjects
STEM cells ,BIOMECHANICS ,MUSCLES ,AUTOMATIC control systems ,BIOMEDICAL engineering - Abstract
Do computational models contribute to progress in mechanobiology? Jacobs and Kelly (in Advances on Modelling in Tissue Engineering, p. 1–14, 2011) suggest that they do, but at the same time propose a limitation in the form of the ‘paradox of validation’, whereby the information needed to validate mechanoregulation theories obviates the need for them in the first place. In this article, the corroboration of theories describing mechanoregulation of tissue differentiation is reviewed. Considering the falsifiability of computational models derived using the theories as a measure of their predictive power, it is shown that the predictive power of some theories is poor and that models based on these theories fall into the ‘paradox of validation’. Week theories for any phenomenon would succumb to such a paradox. We argue that mechanobiology needs theories that can have more potentially falsifying experiments and that perhaps the discipline does suffer from theories that are a priori designed to minimise falsifiability. However, several theories do have predictive power beyond the data used to validate them, so a paradox of validation should disappear as the subject develops. [ABSTRACT FROM PUBLISHER]
- Published
- 2015
- Full Text
- View/download PDF
47. Mechanically induced osteogenic lineage commitment of stem cells.
- Author
-
Chen, Julia C. and Jacobs, Christopher R.
- Subjects
STEM cells ,OSTEOBLASTS ,BONE cells ,CELL adhesion ,TRANSCRIPTION factors ,FOCAL adhesion kinase ,MITOGEN-activated protein kinases - Abstract
Bones adapt to accommodate the physical forces they experience through changes in architecture and mass. Stem cells differentiate into bone-forming osteoblasts, and mechanical stimulation is involved in this process. Various studies have applied controlled mechanical stimulation to stem cells and investigated the effects on osteogenic lineage commitment. These studies demonstrate that physical stimuli can induce osteogenic lineage commitment. Tension, fluid shear stress, substrate material properties, and cell shape are all factors that influence osteogenic differentiation. In particular, the level of tension is important. Also, rigid substrates with stiffness similar to collagenous bone induce osteogenic differentiation, while softer substrates induce other lineages. Finally, cells allowed to adhere over a larger area are able to differentiate towards the osteogenic lineage while cells adhering to a smaller area are restricted to the adipogenic lineage. Stem cells are able to sense their mechanical environments through various mechanosensors, including the cytoskeleton, focal adhesions, and primary cilia. The cytoskeleton provides a structural frame for the cell, and myosin interacts with actin to generate cytoskeletal tension, which is important for mechanically induced osteogenesis of stem cells. Adapter proteins link the cytoskeleton to integrins, which attach the cell to the substrate, forming a focal adhesion. A variety of signaling proteins are also associated with focal adhesions. Forces are transmitted to the substrate at these sites, and an intact focal adhesion is important for mechanically induced osteogenesis. The primary cilium is a single, immotile, antenna-like structure that extends from the cell into the extracellular space. It has emerged as an important signaling center, acting as a microdomain to facilitate biochemical signaling. Mechanotransduction is the process by which physical stimuli are converted into biochemical responses. When potential mechanosensors are disrupted, the activities of components of mechanotransduction pathways are also inhibited, preventing mechanically induced osteogenesis. Calcium, mitogen-activated protein kinase/extracellular signal-regulated kinase, Wnt, Yes-associated protein/transcriptional coactivator with PDZ-binding motif and RhoA/Rho kinase signaling are some of the mechanotransduction pathways proposed to be important. In this review, types of mechanical stimuli, mechanosensors, and key pathways involved in mechanically induced osteogenesis of stem cells are discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
48. Hypoxia promotes chondrogenesis in rat mesenchymal stem cells: A role for AKT and hypoxia-inducible factor (HIF)-1α.
- Author
-
Kanichai, Manoj, Ferguson, Damien, Prendergast, Patrick J., and Campbell, Veronica A.
- Subjects
STEM cells ,CHONDROGENESIS ,TISSUE engineering ,GROWTH factors ,CELL differentiation - Abstract
Mesenchymal stem cells (MSCs) are multipotent cells capable of developing along the chondrogenic, osteogenic and adipogenic lineages. As such, they have received interest as a potential cell source for tissue engineering strategies. Cartilage is an avascular tissue and thus resides in a microenvironment with reduced oxygen tension. The aim of this study was to examine the effect of a low oxygen environment on MSC differentiation along the chondrogenic route. In MSCs exposed to chondrogenic growth factors, transforming growth factor-β and dexamethasone, in a hypoxic environment (2% oxygen), the induction of collagen II expression and proteoglygan deposition was significantly greater than that observed when cells were exposed to the chondrogenic growth factors under normoxic (20% oxygen) conditions. The transcription factor, hypoxia-inducible factor-1α (HIF-1α), is a crucial mediator of the cellular response to hypoxia. Following exposure of MSCs to hypoxia (2% oxygen), HIF-1α translocated from the cytosol to the nucleus and bound to its target DNA consensus sequence. Similarly, hypoxia evoked an increase in phosphorylation of both AKT and p38 mitogen activated protein kinase, upstream of HIF-1α activation. Furthermore, the PI3 kinase/AKT inhibitor, LY294002, and p38 inhibitor, SB 203580, prevented the hypoxia-mediated stabilisation of HIF-1α. To assess the role of HIF-1α in the hypoxia-induced increase in chondrogenesis, we employed an siRNA knockdown approach. In cells exposed to HIF-1α siRNA, the hypoxia-induced enhancement of chondrogenesis, as evidenced by upregulation of collagen II, sox-9 and proteoglycan deposition, was absent. This provides evidence for HIF-1α being a key mediator of the beneficial effect of a low oxygen environment on chondrogenesis. J. Cell. Physiol. 216: 708–715, 2008, © 2008 Wiley-Liss, Inc. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
49. Effects of Cyclic Mechanical Stretching on the mRNA Expression of Tendon/Ligament-Related and Osteoblast-Specific Genes in Human Mesenchymal Stem Cells.
- Author
-
Chen, Yi-Jane, Huang, Chien-Hsun, Lee, I-Chi, Lee, Yu-Tsang, Chen, Min-Huey, and Young, Tai-Horng
- Subjects
BONES ,STEM cells ,MESENCHYME ,TENDONS ,MESSENGER RNA - Abstract
The purpose of this study was to explore the influences of cyclic mechanical stretching on the mRNA expressions of tendon/ligament-related and osteoblast-specific marker genes in human MSCs seeded onto a collagen type I-coated surface. The stretch-induced mRNA expressions of mesenchymal stem cell protein (MSCP), matrix metalloproteinase-3 (MMP-3), and marker genes related to tendon/ligament cells (type I collagen, type III collagen, and tenascin-C) and those typical of osteoblasts (core binding factor alpha 1 (Cbfa1), alkaline phosphatase (ALP), and osteocalcin (OCN)) were analyzed by quantitative real-time PCR. The results revealed significant downregulation of MSCP and upregulation of MMP-3 genes in MSCs subjected to mechanical loading, regardless of the magnitude of the stretching (high or low). Moreover, the typical marker genes of the osteoblast lineage were upregulated by low-magnitude stretching, whereas tendon/ligament-related genes were upregulated by high-magnitude stretching for a long period. Cbfa1 and ALP were upregulated starting as early at 8 hr, followed by a downward trend and no significant change in expression at the other time points. The mRNA expressions of type I collagen, type III collagen, and tenascin-C significantly increased in MSCs subjected to 10% stretching for 48 hr, and this effect still existed after the stretched cells had rested for 48 hr. This study demonstrated the effect of cyclic mechanical stretching on differential transcription of marker genes related to different cell lineages. Low-magnitude stretching increased mRNA expressions of Cbfa1 and ALP and was possibly involved in the early osteoblastic differentiation of MSCs, whereas high-magnitude stretching upregulated the mRNA expressions of tendon/ligament-related genes. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
50. Physical manipulation of calcium oscillations facilitates osteodifferentiation of human mesenchymal stem cells.
- Author
-
Shan Sun, Yaoming Liu, Lipsky, Samantha, and Cho, Michael
- Subjects
CALCIUM ,STEM cells ,MESENCHYME ,CELL differentiation ,ELECTRIC stimulation - Abstract
The role of cytosolic calcium oscillation has long been recognized in the regulation of cellular and molecular interactions. Information embedded in calcium oscillation can provide molecular cues for cell behavior such as cell differentiation. Although calcium dynamics are versatile and likely to depend on the cell type, the calcium dynamics in human mesenchymal stem cells (hMSCs) and its role in differentiation are yet to be fully elucidated. In the present study we characterized the calcium oscillation profiles in hMSCs before and after subjecting the cells to the osteoinductive factors. Our findings indicate that the calcium spikes decreased rapidly with osteodifferentiation to a level observed in terminally differentiated human osteoblasts. In addition, the calcium oscillations appear to serve as a bidirectional signal during hMSC differentiation. While an altered calcium oscillation pattern may be an indicator for hMSC differentiation, it is also likely to be involved in directing hMSC differentiation. Treatment of hMSCs with a noninvasive electrical stimulation, for example, not only altered the calcium oscillations but also facilitated osteodifferentiation. Regulation of calcium oscillation by external physical stimulation could amplify hMSC differentiation into a tissue-specific lineage and may offer an alternate biotechnology to harness the unique properties of stem cells. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.