1
|
Funke S, Wiggenhauser PS, Grundmeier A, Taha S, Fuchs B, Birt A, Koban K, Giunta RE, Kuhlmann C. Aspirin Stimulates the Osteogenic Differentiation of Human Adipose Tissue-Derived Stem Cells In Vitro. Int J Mol Sci 2024; 25:7690. [PMID: 39062933 PMCID: PMC11277042 DOI: 10.3390/ijms25147690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
This study investigates the impact of acetylsalicylic acid (ASA), also known as aspirin, on adipose tissue-derived stem cells (ASCs), aiming to elucidate its dose-dependent effects on morphology, viability, proliferation, and osteogenic differentiation. Isolated and characterized human ASCs were exposed to 0 µM, 100 µM, 200 µM, 400 µM, 800 µM, 1000 µM, 10,000 µM, and 16,000 µM of ASA in vitro. Cell morphology, viability, and proliferation were evaluated with fluorescent live/dead staining, alamarBlue viability reagent, and CyQUANT® cell proliferation assay, respectively. Osteogenic differentiation under stimulation with 400 µM or 1000 µM of ASA was assessed with alizarin red staining and qPCR of selected osteogenic differentiation markers (RUNX2, SPP1, ALPL, BGLAP) over a 3- and 21-day-period. ASA doses ≤ 1000 µM showed no significant impact on cell viability and proliferation. Live/dead staining revealed a visible reduction in viable cell confluency for ASA concentrations ≥ 1000 µM. Doses of 10,000 µM and 16,000 µM of ASA exhibited a strong cytotoxic and anti-proliferative effect in ASCs. Alizarin red staining revealed enhanced calcium accretion under the influence of ASA, which was macro- and microscopically visible and significant for 1000 µM of ASA (p = 0.0092) in quantification if compared to osteogenic differentiation without ASA addition over a 21-day-period. This enhancement correlated with a more pronounced upregulation of osteogenic markers under ASA exposure (ns). Our results indicate a stimulatory effect of 1000 µM of ASA on the osteogenic differentiation of ASCs. Further research is needed to elucidate the precise molecular mechanisms underlying this effect; however, this discovery suggests promising opportunities for enhancing bone tissue engineering with ASCs as cell source.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Constanze Kuhlmann
- Division of Hand Surgery, Plastic Surgery and Aesthetic Surgery, University Hospital, LMU Munich, Ziemssenstraße 5, 80336 Munich, Germany; (S.F.); (P.S.W.); (A.G.); (S.T.); (B.F.); (A.B.); (K.K.); (R.E.G.)
| |
Collapse
|
2
|
Papadopoulos KS, Piperi C, Korkolopoulou P. Clinical Applications of Adipose-Derived Stem Cell (ADSC) Exosomes in Tissue Regeneration. Int J Mol Sci 2024; 25:5916. [PMID: 38892103 PMCID: PMC11172884 DOI: 10.3390/ijms25115916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) are mesenchymal stem cells with a great potential for self-renewal and differentiation. Exosomes derived from ADSCs (ADSC-exos) can imitate their functions, carrying cargoes of bioactive molecules that may affect specific cellular targets and signaling processes. Recent evidence has shown that ADSC-exos can mediate tissue regeneration through the regulation of the inflammatory response, enhancement of cell proliferation, and induction of angiogenesis. At the same time, they may promote wound healing as well as the remodeling of the extracellular matrix. In combination with scaffolds, they present the future of cell-free therapies and promising adjuncts to reconstructive surgery with diverse tissue-specific functions and minimal adverse effects. In this review, we address the main characteristics and functional properties of ADSC-exos in tissue regeneration and explore their most recent clinical application in wound healing, musculoskeletal regeneration, dermatology, and plastic surgery as well as in tissue engineering.
Collapse
Affiliation(s)
- Konstantinos S. Papadopoulos
- Department of Plastic and Reconstructive Surgery, 401 General Military Hospital of Athens, 11525 Athens, Greece;
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527 Athens, Greece
| | - Penelope Korkolopoulou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
3
|
Zawrzykraj M, Deptuła M, Kondej K, Tymińska A, Pikuła M. The effect of chemotherapy and radiotherapy on stem cells and wound healing. Current perspectives and challenges for cell-based therapies. Biomed Pharmacother 2023; 168:115781. [PMID: 39491418 DOI: 10.1016/j.biopha.2023.115781] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/05/2024] Open
Abstract
Cancers are part of the group of diseases that carry a high mortality rate. According to World Health Organization in 2020 reported 10 million deaths due to cancers. Treatment of oncological patients is focused on chemotherapeutic agents, radiology, or immunology. Surgical interventions are also an important aspect of treatment. The above methods contribute to saving the patients' health and lives. However, cancer treatment possesses side effects. Commonly observed complications are hair loss, mucositis, nausea, diarrhea, or various skin damage. To improve the quality of medical care for cancer patients, new methods of reducing side effects are sought. Strategies include the use of stem cells (SCs). Due to unlimited proliferation potential and differentiating abilities, SCs are used in the treatment of many disease entities, including wounds. One of the most used types of stem cells supposed adipose-derived mesenchymal stromal cells (AD-MSCs). Clinical trials confirm the application of AD-MSCs in wound healing. Furthermore, in vivo studies considered the utilization of AD-MSCs in radiation injury. The use of stem cells in cancer treatment still involves many questions, such as the impact of treatment on SCs' condition and oncological safety. However, development in regenerative medicine research may contribute to the use of stem cells in personalized medicine, customized for the patient. This could represent a breakthrough step in preventing the side effects of cancer therapies, including chronic wounds.
Collapse
Affiliation(s)
| | - Milena Deptuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Poland
| | - Karolina Kondej
- Department of Plastic Surgery, Medical University of Gdansk, Poland
| | - Agata Tymińska
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Poland
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Poland.
| |
Collapse
|
4
|
Xu X, Zhao L, Terry PD, Chen J. Reciprocal Effect of Environmental Stimuli to Regulate the Adipogenesis and Osteogenesis Fate Decision in Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs). Cells 2023; 12:1400. [PMID: 37408234 PMCID: PMC10216952 DOI: 10.3390/cells12101400] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
Mesenchymal stem cells derived from bone marrow (BM-MSCs) can differentiate into adipocytes and osteoblasts. Various external stimuli, including environmental contaminants, heavy metals, dietary, and physical factors, are shown to influence the fate decision of BM-MSCs toward adipogenesis or osteogenesis. The balance of osteogenesis and adipogenesis is critical for the maintenance of bone homeostasis, and the interruption of BM-MSCs lineage commitment is associated with human health issues, such as fracture, osteoporosis, osteopenia, and osteonecrosis. This review focuses on how external stimuli shift the fate of BM-MSCs towards adipogenesis or osteogenesis. Future studies are needed to understand the impact of these external stimuli on bone health and elucidate the underlying mechanisms of BM-MSCs differentiation. This knowledge will inform efforts to prevent bone-related diseases and develop therapeutic approaches to treat bone disorders associated with various pathological conditions.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Ling Zhao
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Paul D. Terry
- Department of Medicine, Graduate School of Medicine, The University of Tennessee, Knoxville, TN 37920, USA;
| | - Jiangang Chen
- Department of Public Health, The University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
5
|
Research progress of stem cell therapy for endometrial injury. Mater Today Bio 2022; 16:100389. [PMID: 36033375 PMCID: PMC9403503 DOI: 10.1016/j.mtbio.2022.100389] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/24/2022] Open
Abstract
Endometrial damage is an important factor leading to infertility and traditional conventional treatments have limited efficacy. As an emerging technology in recent years, stem cell therapy has provided new hope for the treatment of this disease. By comparing the advantages of stem cells from different sources, it is believed that menstrual blood endometrial stem cells have a good application prospect as a new source of stem cells. However, the clinical utility of stem cells is still limited by issues such as colonization rates, long-term efficacy, tumor formation, and storage and transportation. This paper summarizes the mechanism by which stem cells repair endometrial damage and clarifies the material basis of their effects from four aspects: replacement of damaged sites, paracrine effects, interaction with growth factors, and other new targets. According to the pathological characteristics and treatment requirements of intrauterine adhesion (IUA), the research work to solve the above problems from the aspects of functional bioscaffold preparation and multi-functional platform construction is also summarized. From the perspective of scaffold materials and component functions, this review will provide a reference for comprehensively optimizing the clinical application of stem cells.
Collapse
|
6
|
Liu Y, Wang X, Wu Q, Pei W, Teo MJ, Chen ZS, Huang C. Application of lignin and lignin-based composites in different tissue engineering fields. Int J Biol Macromol 2022; 222:994-1006. [DOI: 10.1016/j.ijbiomac.2022.09.267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/17/2022] [Accepted: 09/28/2022] [Indexed: 12/17/2022]
|
7
|
Voynova E, Kulebyakin K, Grigorieva O, Novoseletskaya E, Basalova N, Alexandrushkina N, Arbatskiy M, Vigovskiy M, Sorokina A, Zinoveva A, Bakhchinyan E, Kalinina N, Akopyan Z, Tkachuk V, Tyurin-Kuzmin P, Efimenko A. Declined adipogenic potential of senescent MSCs due to shift in insulin signaling and altered exosome cargo. Front Cell Dev Biol 2022; 10:1050489. [PMID: 36467400 PMCID: PMC9714334 DOI: 10.3389/fcell.2022.1050489] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/02/2022] [Indexed: 11/19/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) maintain cellular homeostasis and regulate tissue renewal and repair both by differentiating into mesodermal lineage, e.g., adipocytes, or managing the functions of differentiated cells. Insulin is a key physiological inducer of MSC differentiation into adipocytes, and disturbances in MSC insulin sensitivity could negatively affect adipose tissue renewal. During aging, regulation and renewal of adipose tissue cells may be disrupted due to the altered insulin signaling and differentiation potential of senescent MSCs, promoting the development of serious metabolic diseases, including metabolic syndrome and obesity. However, the potential mechanisms mediating the dysfunction of adipose-derived senescent MSC remains unclear. We explored whether aging could affect the adipogenic potential of human adipose tissue-derived MSCs regulated by insulin. Age-associated senescent MSCs (isolated from donors older than 65 years) and MSCs in replicative senescence (long-term culture) were treated by insulin to induce adipogenic differentiation, and the efficiency of the process was compared to MSCs from young donors. Insulin-dependent signaling pathways were explored in these cells. We also analyzed the involvement of extracellular vesicles secreted by MSCs (MSC-EVs) into the regulation of adipogenic differentiation and insulin signaling of control and senescent cells. Also the microRNA profiles of MSC-EVs from aged and young donors were compared using targeted PCR arrays. Both replicatively and chronologically senescent MSCs showed a noticeably decreased adipogenic potential. This was associated with insulin resistance of MSCs from aged donors caused by the increase in the basal level of activation of crucial insulin-dependent intracellular effectors ERK1/2 and Akt. To assess the impact of the paracrine cross-talk of MSCs, we analyzed microRNAs profile differences in MSC-EVs and revealed that senescent MSCs produced EVs with increased content of miRNAs targeting components of insulin-dependent signaling cascade PTEN, MAPK1, GAREM1 and some other targets. We also confirmed these data by differentiation of control MSCs in the presence of EVs from senescent cells and vice versa. Thus, aging attenuated the adipogenic potential of MSCs due to autocrine or paracrine-dependent induction of insulin resistance associated with the specific changes in MSC-EV cargo.
Collapse
Affiliation(s)
- Elizaveta Voynova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia,*Correspondence: Elizaveta Voynova, ; Pyotr Tyurin-Kuzmin, ; Anastasia Efimenko,
| | - Konstantin Kulebyakin
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia,Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Grigorieva
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina Novoseletskaya
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Natalia Basalova
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Natalia Alexandrushkina
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Mikhail Arbatskiy
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Maxim Vigovskiy
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Anna Sorokina
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Anna Zinoveva
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | | | - Natalia Kalinina
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Zhanna Akopyan
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Vsevolod Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Pyotr Tyurin-Kuzmin
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia,*Correspondence: Elizaveta Voynova, ; Pyotr Tyurin-Kuzmin, ; Anastasia Efimenko,
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia,*Correspondence: Elizaveta Voynova, ; Pyotr Tyurin-Kuzmin, ; Anastasia Efimenko,
| |
Collapse
|
8
|
Gundacker C, Audouze K, Widhalm R, Granitzer S, Forsthuber M, Jornod F, Wielsøe M, Long M, Halldórsson TI, Uhl M, Bonefeld-Jørgensen EC. Reduced Birth Weight and Exposure to Per- and Polyfluoroalkyl Substances: A Review of Possible Underlying Mechanisms Using the AOP-HelpFinder. TOXICS 2022; 10:toxics10110684. [PMID: 36422892 PMCID: PMC9699222 DOI: 10.3390/toxics10110684] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 05/14/2023]
Abstract
Prenatal exposure to per- and polyfluorinated substances (PFAS) may impair fetal growth. Our knowledge of the underlying mechanisms is incomplete. We used the Adverse Outcome Pathway (AOP)-helpFinder tool to search PubMed for studies published until March 2021 that examined PFAS exposure in relation to birth weight, oxidative stress, hormones/hormone receptors, or growth signaling pathways. Of these 1880 articles, 106 experimental studies remained after abstract screening. One clear finding is that PFAS are associated with oxidative stress in in vivo animal studies and in vitro studies. It appears that PFAS-induced reactive-oxygen species (ROS) generation triggers increased peroxisome proliferator-activated receptor (PPAR)γ expression and activation of growth signaling pathways, leading to hyperdifferentiation of pre-adipocytes. Fewer proliferating pre-adipocytes result in lower adipose tissue weight and in this way may reduce birth weight. PFAS may also impair fetal growth through endocrine effects. Estrogenic effects have been noted in in vivo and in vitro studies. Overall, data suggest thyroid-damaging effects of PFAS affecting thyroid hormones, thyroid hormone gene expression, and histology that are associated in animal studies with decreased body and organ weight. The effects of PFAS on the complex relationships between oxidative stress, endocrine system function, adipogenesis, and fetal growth should be further explored.
Collapse
Affiliation(s)
- Claudia Gundacker
- Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: ; Tel.: +43-1-40160-56503
| | - Karine Audouze
- Unit T3S, Université Paris Cité, Inserm U1124, 75006 Paris, France
| | - Raimund Widhalm
- Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria
| | - Sebastian Granitzer
- Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin Forsthuber
- Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria
| | - Florence Jornod
- Unit T3S, Université Paris Cité, Inserm U1124, 75006 Paris, France
| | - Maria Wielsøe
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark
| | - Manhai Long
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark
| | - Thórhallur Ingi Halldórsson
- Faculty of Food Science and Nutrition, University of Iceland, 102 Reykjavík, Iceland
- Department of Epidemiology Research, Statens Serum Institut, 2300 Copenhagen, Denmark
| | - Maria Uhl
- Environment Agency Austria, 1090 Vienna, Austria
| | - Eva Cecilie Bonefeld-Jørgensen
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark
- Greenland Center for Health Research, Greenland University, Nuuk 3905, Greenland
| |
Collapse
|
9
|
Long C, Wang J, Gan W, Qin X, Yang R, Chen X. Therapeutic potential of exosomes from adipose-derived stem cells in chronic wound healing. Front Surg 2022; 9:1030288. [PMID: 36248361 PMCID: PMC9561814 DOI: 10.3389/fsurg.2022.1030288] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic wound healing remains a challenging medical problem affecting society, which urgently requires anatomical and functional solutions. Adipose-derived stem cells (ADSCs), mesenchymal stem cells with self-renewal and multiple differentiation ability, play essential roles in wound healing and tissue regeneration. The exosomes from ADSCs (ADSC-EXOs) are extracellular vesicles that are essential for communication between cells. ADSC-EXOs release various bioactive molecules and subsequently restore tissue homeostasis and accelerate wound healing, by promoting various stages of wound repair, including regulating the inflammatory response, promoting wound angiogenesis, accelerating cell proliferation, and modulating wound remodeling. Compared with ADSCs, ADSC-EXOs have the advantages of avoiding ethical issues, being easily stored, and having high stability. In this review, a literature search of PubMed, Medline, and Google Scholar was performed for articles before August 1, 2022 focusing on exosomes from ADSCs, chronic wound repair, and therapeutic potential. This review aimed to provide new therapeutic strategies to help investigators explore how ADSC-EXOs regulate intercellular communication in chronic wounds.
Collapse
Affiliation(s)
- Chengmin Long
- Guangdong Medical University, Zhanjiang, China
- Department of Burn Surgery and Skin Regeneration, the First People’s Hospital of Foshan, Foshan, China
| | - Jingru Wang
- Department of Burn Surgery and Skin Regeneration, the First People’s Hospital of Foshan, Foshan, China
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Wenjun Gan
- Guangdong Medical University, Zhanjiang, China
- Department of Burn Surgery and Skin Regeneration, the First People’s Hospital of Foshan, Foshan, China
| | - Xinchi Qin
- Department of Burn Surgery and Skin Regeneration, the First People’s Hospital of Foshan, Foshan, China
- Zunyi Medical University, Zhuhai, China
| | - Ronghua Yang
- Guangdong Medical University, Zhanjiang, China
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
- Correspondence: Xiaodong Chen Ronghua Yang a_hwa991316 @163.com
| | - Xiaodong Chen
- Guangdong Medical University, Zhanjiang, China
- Department of Burn Surgery and Skin Regeneration, the First People’s Hospital of Foshan, Foshan, China
- Correspondence: Xiaodong Chen Ronghua Yang a_hwa991316 @163.com
| |
Collapse
|
10
|
Ghasroldasht MM, Mastrogiacomo M, Akbarian F, Rezaeian A. Polyurethane and polyurethane/hydroxyapatite scaffold in a three-dimensional culture system. Cell Biol Int 2022; 46:2041-2049. [PMID: 35971683 DOI: 10.1002/cbin.11878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 11/10/2022]
Abstract
Designing a new scaffold with an optimal ability of osteogenesis differentiation is a significant step bone tissue engineering along with the growing demands for bone craft in recent decades. Herein, we used Polyurethane (PU), a novel biocompatible and flexible polymer, and Hydroxyapatite (HA), the major component of human hard tissues matrix for developing new scaffolds and analyzing the in vitro osteogenic differentiation potential of human adipose-derived mesenchymal stem cells (Ad-MSCs) in basal and induction media. Gene expression analysis was performed to evaluate the expression level of four osteogenic differentiation genes. MTT assays were also done to assess the attachment and proliferation of the cells after 7 and 21 days of seeding to scaffolds. The expression level of RUNX2 was increased in seeded cells on PU/HA scaffolds compared with the PU. Cellular adhesion and proliferation of the Ad-MSCs were higher in PU/HA than PU scaffolds according to the histology analysis. The PU and PU/HA scaffolds supported the attachment, proliferation, and differentiation of Ad-MSCs, and they are suitable candidates for producing constructs in bone regeneration. However, further in-vitro and in-vivo studies on these scaffolds are needed to introduce an appropriate candidate for clinical bone regeneration.
Collapse
Affiliation(s)
| | | | - Fahimeh Akbarian
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefeh Rezaeian
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
11
|
TGFβ-Treated Placenta-Derived Mesenchymal Stem Cells Selectively Promote Anti-Adipogenesis in Thyroid-Associated Ophthalmopathy. Int J Mol Sci 2022; 23:ijms23105603. [PMID: 35628410 PMCID: PMC9145654 DOI: 10.3390/ijms23105603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023] Open
Abstract
Orbital fibroblasts (OFs) in thyroid-associated ophthalmopathy (TAO) are differentiated from pre-adipocytes and mature adipocytes; increased lipid and fat expansion are the major characteristics of ophthalmic manifestations. Human placental mesenchymal stem cells (hPMSCs) were reported to immunomodulate pathogenesis and suppress adipogenesis in TAO OFs. Here, we prepared transforming growth factor β (TGFβ, 20 ng/mL)-treated hPMSCs (TGFβ-hPMSCs) in order to enhance anti-adipogenic effects in vitro and in TAO mice. TAO OFs were grown in a differentiation medium and then co-cultured with hPMSCs or TGFβ-hPMSCs. TAO OFs were analyzed via quantitative real-time polymerase chain reaction, Oil red O staining, and western blotting. The results showed that TGFβ-hPMSCs reduced the expression of adipogenic, lipogenic, and fibrotic genes better than hPMSCs in TAO OFs. Moreover, the adipose area decreased more in TAO mice injected with TGFβ-hPMSCs compared to those injected with hPMSCs or a steroid. Further, TGFβ-hPMSCs inhibited inflammation as effectively as a steroid. In conclusion, TGFβ-hPMSCs suppressed adipogenesis and lipogenesis in vitro and in TAO mice, and the effects were mediated by the SMAD 2/3 pathways. Furthermore, TGFβ-hPMSCs exhibited anti-inflammatory and anti-fibrotic functions, which suggests that they could be a new and safe method to promote the anti-adipogenic function of hPMSCs to treat TAO patients.
Collapse
|
12
|
Yao W, Shi L, Zhang Y, Dong H, Zhang Y. Mesenchymal stem/stromal cell therapy for COVID-19 pneumonia: potential mechanisms, current clinical evidence, and future perspectives. Stem Cell Res Ther 2022; 13:124. [PMID: 35321737 PMCID: PMC8942612 DOI: 10.1186/s13287-022-02810-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/07/2022] [Indexed: 12/20/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread into more than 200 countries and infected approximately 203 million people globally. COVID-19 is associated with high mortality and morbidity in some patients, and this disease still does not have effective treatments with reproducibly appreciable outcomes. One of the leading complications associated with COVID-19 is acute respiratory distress syndrome (ARDS); this is an anti-viral host inflammatory response, and it is usually caused by a cytokine storm syndrome which may lead to multi-organ failure and death. Currently, COVID-19 patients are treated with approaches that mostly fall into two major categories: immunomodulators, which promote the body's fight against viruses efficiently, and antivirals, which slow or stop viruses from multiplying. These treatments include a variety of novel therapies that are currently being tested in clinical trials, including serum, IL-6 antibody, and remdesivir; however, the outcomes of these therapies are not consistently appreciable and remain a subject of debate. Mesenchymal stem/stromal cells (MSCs), the multipotent stem cells that have previously been used to treat viral infections and various respiratory diseases such as ARDS exhibit immunomodulatory properties and can ameliorate tissue damage. Given that SARS-CoV-2 targets the immune system and causes tissue damage, it is presumable that MSCs are being explored to treat COVID-19 patients. This review summarizes the potential mechanisms of action of MSC therapy, progress of MSC, and its related products in clinical trials for COVID-19 therapy based on the outcomes of these clinical studies.
Collapse
Affiliation(s)
- Weiqi Yao
- Department of Hematology, Union Hospital, Tong Ji Medical College, Hua Zhong University of Science and Technology, Hubei, China
- State Industrial Base for Stem Cell Engineering Products, No. 12 Meiyuan Road, Tianjin, 300384, China
- Hubei Engineering Research Center for Human Stem Cell Preparation, Application and Resource Preservation, Wuhan, China
| | - Lei Shi
- Department of Infectious Diseases, Fifth Medical Center of Chinese, PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yun Zhang
- State Industrial Base for Stem Cell Engineering Products, No. 12 Meiyuan Road, Tianjin, 300384, China
- Tianjin Key Laboratory for Stem Cell and Regenerative Medicine, Tianjin, China
| | - Haibo Dong
- Hubei Engineering Research Center for Human Stem Cell Preparation, Application and Resource Preservation, Wuhan, China
- Wuhan Optics Valley VCANBIO Cell & Gene Technology Co., Ltd., Hubei, China
| | - Yu Zhang
- State Industrial Base for Stem Cell Engineering Products, No. 12 Meiyuan Road, Tianjin, 300384, China.
- Hubei Engineering Research Center for Human Stem Cell Preparation, Application and Resource Preservation, Wuhan, China.
- Tianjin Key Laboratory for Stem Cell and Regenerative Medicine, Tianjin, China.
- Tianjin Key Laboratory for Blood Cell Therapy Technology, Tianjin, China.
| |
Collapse
|
13
|
Examination of the Quality of Particulate and Filtered Mandibular Bone Chips for Oral Implants: An In Vitro Study. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12042031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
(1) Background: Autologous bone is supposed to contain vital cells that might improve the osseointegration of dental implants. The aim of this study was to investigate particulate and filtered bone chips collected during oral surgery intervention with respect to their osteogenic potential and the extent of microbial contamination to evaluate its usefulness for jawbone reconstruction prior to implant placement. (2) Methods: Cortical and cortical-cancellous bone chip samples of 84 patients were collected. The stem cell character of outgrowing cells was characterized by expression of CD73, CD90 and CD105, followed by osteogenic differentiation. The degree of bacterial contamination was determined by Gram staining, catalase and oxidase tests and tests to evaluate the genera of the found bacteria (3) Results: Pre-surgical antibiotic treatment of the patients significantly increased viability of the collected bone chip cells. No significant difference in plasticity was observed between cells isolated from the cortical and cortical-cancellous bone chip samples. Thus, both types of bone tissue can be used for jawbone reconstruction. The osteogenic differentiation was independent of the quantity and quality of the detected microorganisms, which comprise the most common bacteria in the oral cavity. (4) Discussion: This study shows that the quality of bone chip-derived stem cells is independent of the donor site and the extent of present common microorganisms, highlighting autologous bone tissue, assessable without additional surgical intervention for the patient, as a useful material for dental implantology.
Collapse
|
14
|
Tonk CH, Shoushrah SH, Babczyk P, El Khaldi-Hansen B, Schulze M, Herten M, Tobiasch E. Therapeutic Treatments for Osteoporosis-Which Combination of Pills Is the Best among the Bad? Int J Mol Sci 2022; 23:1393. [PMID: 35163315 PMCID: PMC8836178 DOI: 10.3390/ijms23031393] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis is a chronical, systemic skeletal disorder characterized by an increase in bone resorption, which leads to reduced bone density. The reduction in bone mineral density and therefore low bone mass results in an increased risk of fractures. Osteoporosis is caused by an imbalance in the normally strictly regulated bone homeostasis. This imbalance is caused by overactive bone-resorbing osteoclasts, while bone-synthesizing osteoblasts do not compensate for this. In this review, the mechanism is presented, underlined by in vitro and animal models to investigate this imbalance as well as the current status of clinical trials. Furthermore, new therapeutic strategies for osteoporosis are presented, such as anabolic treatments and catabolic treatments and treatments using biomaterials and biomolecules. Another focus is on new combination therapies with multiple drugs which are currently considered more beneficial for the treatment of osteoporosis than monotherapies. Taken together, this review starts with an overview and ends with the newest approaches for osteoporosis therapies and a future perspective not presented so far.
Collapse
Affiliation(s)
- Christian Horst Tonk
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Sarah Hani Shoushrah
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Patrick Babczyk
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Basma El Khaldi-Hansen
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Monika Herten
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| |
Collapse
|
15
|
Application of Stem Cell Therapy for ACL Graft Regeneration. Stem Cells Int 2021; 2021:6641818. [PMID: 34381504 PMCID: PMC8352687 DOI: 10.1155/2021/6641818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/19/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Graft regeneration after anterior cruciate ligament (ACL) reconstruction surgery is a complex three-stage process, which usually takes a long duration and often results in fibrous scar tissue formation that exerts a detrimental impact on the patients' prognosis. Hence, as a regeneration technique, stem cell transplantation has attracted increasing attention. Several different stem cell types have been utilized in animal experiments, and almost all of these have shown good capacity in improving tendon-bone regeneration. Various differentiation inducers have been widely applied together with stem cells to enhance specific lineage differentiation, such as recombinant gene transfection, growth factors, and biomaterials. Among the various different types of stem cells, bone marrow-derived mesenchymal stem cells (BMSCs) have been investigated the most, while ligament stem progenitor cells (LDSCs) have demonstrated the best potential in generating tendon/ligament lineage cells. In the clinic, 4 relevant completed trials have been reported, but only one trial with BMSCs showed improved outcomes, while 5 relevant trials are still in progress. This review describes the process of ACL graft regeneration after implantation and summarizes the current application of stem cells from bench to bedside, as well as discusses future perspectives in this field.
Collapse
|
16
|
Shoushrah SH, Transfeld JL, Tonk CH, Büchner D, Witzleben S, Sieber MA, Schulze M, Tobiasch E. Sinking Our Teeth in Getting Dental Stem Cells to Clinics for Bone Regeneration. Int J Mol Sci 2021; 22:6387. [PMID: 34203719 PMCID: PMC8232184 DOI: 10.3390/ijms22126387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Dental stem cells have been isolated from the medical waste of various dental tissues. They have been characterized by numerous markers, which are evaluated herein and differentiated into multiple cell types. They can also be used to generate cell lines and iPSCs for long-term in vitro research. Methods for utilizing these stem cells including cellular systems such as organoids or cell sheets, cell-free systems such as exosomes, and scaffold-based approaches with and without drug release concepts are reported in this review and presented with new pictures for clarification. These in vitro applications can be deployed in disease modeling and subsequent pharmaceutical research and also pave the way for tissue regeneration. The main focus herein is on the potential of dental stem cells for hard tissue regeneration, especially bone, by evaluating their potential for osteogenesis and angiogenesis, and the regulation of these two processes by growth factors and environmental stimulators. Current in vitro and in vivo publications show numerous benefits of using dental stem cells for research purposes and hard tissue regeneration. However, only a few clinical trials currently exist. The goal of this review is to pinpoint this imbalance and encourage scientists to pick up this research and proceed one step further to translation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig- Strasse. 20, 53359 Rheinbach, Germany; (S.H.S.); (J.L.T.); (C.H.T.); (D.B.); (S.W.); (M.A.S.); (M.S.)
| |
Collapse
|
17
|
Tan L, Tran L, Ferreyra S, Moran JA, Skovgaard Z, Trujillo A, ibili E, Zhao Y. Downregulation of SUV39H1 and CITED2 Exerts Additive Effect on Promoting Adipogenic Commitment of Human Mesenchymal Stem Cells. Stem Cells Dev 2021; 30:485-501. [PMID: 33691475 PMCID: PMC8106253 DOI: 10.1089/scd.2020.0190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/10/2021] [Indexed: 11/12/2022] Open
Abstract
Human adipogenesis is the process through which uncommitted human mesenchymal stem cells (hMSCs) differentiate into adipocytes. Through a siRNA-based high-throughput screen that identifies adipogenic regulators whose expression knockdown leads to enhanced adipogenic differentiation of hMSCs, two new regulators, SUV39H1, a histone methyltransferase that catalyzes H3K9Me3, and CITED2, a CBP/p300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 2 were uncovered. Both SUV39H1 and CITED2 are normally downregulated during adipogenic differentiation of hMSCs. Further expression knockdown induced by siSUV39H1 or siCITED2 at the adipogenic initiation stage significantly enhanced adipogenic differentiation of hMSCs as compared with siControl treatment, with siSUV39H1 acting by both accelerating fat accumulation in individual adipocytes and increasing the total number of committed adipocytes, whereas siCITED2 acting predominantly by increasing the total number of committed adipocytes. In addition, both siSUV39H1 and siCITED2 were able to redirect hMSCs to undergo adipogenic differentiation in the presence of osteogenic inducing media, which normally only induces osteogenic differentiation of hMSCs in the absence of siSUV39H1 or siCITED2. Interestingly, simultaneous knockdown of both SUV39H1 and CITED2 resulted in even greater levels of adipogenic differentiation of hMSCs and expression of CEBPα and PPARγ, two master regulators of adipogenesis, as compared with those elicited by single gene knockdown. Furthermore, the effects of co-knockdown were equivalent to the additive effect of individual gene knockdown. Taken together, this study demonstrates that SUV39H1 and CITED2 are both negative regulators of human adipogenesis, and downregulation of both genes exerts an additive effect on promoting adipogenic differentiation of hMSCs through augmented commitment.
Collapse
Affiliation(s)
- Lun Tan
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Linh Tran
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Stephanie Ferreyra
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Jose A. Moran
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Zachary Skovgaard
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Amparo Trujillo
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Esra ibili
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| | - Yuanxiang Zhao
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, USA
| |
Collapse
|
18
|
Collagen type I promotes osteogenic differentiation of amniotic membrane-derived mesenchymal stromal cells in basal and induction media. Biosci Rep 2021; 40:227060. [PMID: 33245097 PMCID: PMC7736623 DOI: 10.1042/bsr20201325] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
Collagen has been widely shown to promote osteogenesis of bone marrow mesenchymal stromal cells (BM-MSCs). Due to the invasive procedure of obtaining BM-MSCs, MSCs from other tissues have emerged as a promising alternative for regenerative therapy. MSCs originated from different sources, exhibiting different differentiation potentials. Therefore, the applicability of collagen type I (COL), combining with amniotic membrane (AM)-MSCs was examined through proliferation and differentiation assays together with the expression of surface markers and genes associated with stemness and differentiation under basal or induction conditions. No increase in cell growth was observed because AM-MSCs might be directed toward spontaneous osteogenesis. This was evidenced by the calcium deposition and elevated expression of osteogenic genes when AM-MSCs were cultured in collagen plate with basal media. Under the osteogenic condition, reciprocal expression of OCN and CEBPA suggested a shift toward adipogenesis. Surprisingly, adipogenic genes were not elevated upon adipogenic induction, although oil droplets deposition was observed. In conclusion, our findings demonstrated that collagen causes spontaneous osteogenesis in AM-MSCs. However, the presence of exogenous inductors could shift the direction of adipo-osteogenic gene regulatory network modulated by collagen.
Collapse
|
19
|
Woods DC, Rodríguez-Ropero F, Wereszczynski J. The Dynamic Influence of Linker Histone Saturation within the Poly-Nucleosome Array. J Mol Biol 2021; 433:166902. [PMID: 33667509 DOI: 10.1016/j.jmb.2021.166902] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 02/08/2023]
Abstract
Linker histones bind to nucleosomes and modify chromatin structure and dynamics as a means of epigenetic regulation. Biophysical studies have shown that chromatin fibers can adopt a plethora of conformations with varying levels of compaction. Linker histone condensation, and its specific binding disposition, has been associated with directly tuning this ensemble of states. However, the atomistic dynamics and quantification of this mechanism remains poorly understood. Here, we present molecular dynamics simulations of octa-nucleosome arrays, based on a cryo-EM structure of the 30-nm chromatin fiber, with and without the globular domains of the H1 linker histone to determine how they influence fiber structures and dynamics. Results show that when bound, linker histones inhibit DNA flexibility and stabilize repeating tetra-nucleosomal units, giving rise to increased chromatin compaction. Furthermore, upon the removal of H1, there is a significant destabilization of this compact structure as the fiber adopts less strained and untwisted states. Interestingly, linker DNA sampling in the octa-nucleosome is exaggerated compared to its mono-nucleosome counterparts, suggesting that chromatin architecture plays a significant role in DNA strain even in the absence of linker histones. Moreover, H1-bound states are shown to have increased stiffness within tetra-nucleosomes, but not between them. This increased stiffness leads to stronger long-range correlations within the fiber, which may result in the propagation of epigenetic signals over longer spatial ranges. These simulations highlight the effects of linker histone binding on the internal dynamics and global structure of poly-nucleosome arrays, while providing physical insight into a mechanism of chromatin compaction.
Collapse
Affiliation(s)
- Dustin C Woods
- Department of Chemistry and the Center for Molecular Study of Condensed Soft Matter, Illinois Institute of Technology, Chicago, IL 60616, United States
| | - Francisco Rodríguez-Ropero
- Department of Physics and the Center for Molecular Study of Condensed Soft Matter, Illinois Institute of Technology, Chicago, IL 60616, United States
| | - Jeff Wereszczynski
- Department of Physics and the Center for Molecular Study of Condensed Soft Matter, Illinois Institute of Technology, Chicago, IL 60616, United States.
| |
Collapse
|
20
|
Regenerative Medicine for Equine Musculoskeletal Diseases. Animals (Basel) 2021; 11:ani11010234. [PMID: 33477808 PMCID: PMC7832834 DOI: 10.3390/ani11010234] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 01/15/2023] Open
Abstract
Simple Summary Lameness due to musculoskeletal disease is the most common diagnosis in equine veterinary practice. Many of these orthopaedic disorders are chronic problems, for which no clinically satisfactory treatment exists. Thus, high hopes are pinned on regenerative medicine, which aims to replace or regenerate cells, tissues, or organs to restore or establish normal function. Some regenerative medicine therapies have already made their way into equine clinical practice mainly to treat tendon injures, tendinopathies, cartilage injuries and degenerative joint disorders with promising but diverse results. This review summarises the current knowledge of commonly used regenerative medicine treatments and critically discusses their use. Abstract Musculoskeletal injuries and chronic degenerative diseases commonly affect both athletic and sedentary horses and can entail the end of their athletic careers. The ensuing repair processes frequently do not yield fully functional regeneration of the injured tissues but biomechanically inferior scar or replacement tissue, causing high reinjury rates, degenerative disease progression and chronic morbidity. Regenerative medicine is an emerging, rapidly evolving branch of translational medicine that aims to replace or regenerate cells, tissues, or organs to restore or establish normal function. It includes tissue engineering but also cell-based and cell-free stimulation of endogenous self-repair mechanisms. Some regenerative medicine therapies have made their way into equine clinical practice mainly to treat tendon injures, tendinopathies, cartilage injuries and degenerative joint disorders with promising results. However, the qualitative and quantitative spatiotemporal requirements for specific bioactive factors to trigger tissue regeneration in the injury response are still unknown, and consequently, therapeutic approaches and treatment results are diverse. To exploit the full potential of this burgeoning field of medicine, further research will be required and is ongoing. This review summarises the current knowledge of commonly used regenerative medicine treatments in equine patients and critically discusses their use.
Collapse
|
21
|
Yamada S, Yassin MA, Schwarz T, Hansmann J, Mustafa K. Induction of osteogenic differentiation of bone marrow stromal cells on 3D polyester-based scaffolds solely by subphysiological fluidic stimulation in a laminar flow bioreactor. J Tissue Eng 2021; 12:20417314211019375. [PMID: 34262684 PMCID: PMC8243246 DOI: 10.1177/20417314211019375] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/04/2021] [Indexed: 01/09/2023] Open
Abstract
The fatal determination of bone marrow mesenchymal stem/stromal cells (BMSC) is closely associated with mechano-environmental factors in addition to biochemical clues. The aim of this study was to induce osteogenesis in the absence of chemical stimuli using a custom-designed laminar flow bioreactor. BMSC were seeded onto synthetic microporous scaffolds and subjected to the subphysiological level of fluid flow for up to 21 days. During the perfusion, cell proliferation was significantly inhibited. There were also morphological changes, with F-actin polymerisation and upregulation of ROCK1. Notably, in BMSC subjected to flow, mRNA expression of osteogenic markers was significantly upregulated and RUNX2 was localised in the nuclei. Further, under perfusion, there was greater deposition of collagen type 1 and calcium onto the scaffolds. The results confirm that an appropriate level of fluid stimuli preconditions BMSC towards the osteoblastic lineage on 3D scaffolds in the absence of chemical stimulation, which highlights the utility of flow bioreactors in bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Mohammed Ahmed Yassin
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Thomas Schwarz
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
| | - Jan Hansmann
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
- Chair of Tissue Engineering and
Regenerative Medicine, University Hospital Würzburg, Germany
- Department Electrical Engineering,
University of Applied Sciences Würzburg-Schweinfurt, Germany
| | - Kamal Mustafa
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| |
Collapse
|
22
|
Piao J, Park JS, Hwang DY, Son Y, Hong HS. Substance P blocks ovariectomy-induced bone loss by modulating inflammation and potentiating stem cell function. Aging (Albany NY) 2020; 12:20753-20777. [PMID: 33109775 PMCID: PMC7655156 DOI: 10.18632/aging.104008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022]
Abstract
Osteoporosis is an age-related disease caused by imbalanced bone remodeling resulting from excessive bone resorption. Osteoporosis is tightly linked with induction of chronic inflammation, which activates osteoclasts and impairs osteoprogenitor in bone marrow. T helper 17 (Th17) cells have been recently recognized as one of major inducers in the pathophysiology of bone loss by secreting IL-17. Thus, modulation of Th17 development is anticipated to affect the progression of osteoporosis. Substance P (SP) is reported to provide anti-inflammatory effects by controlling immune cell profile and also, promote restoration of damaged stem cell niches—the bone marrow—by repopulating BMSCs or potentiating its paracrine ability. This study aimed to explore the therapeutic effects of systemically injected SP on ovariectomy (OVX)-induced osteoporosis. Resultantly, SP injection obviously blocked OVX-induced impairment of bone microarchitecture and reduction of the mineral density. In osteoporotic condition, SP could ameliorate chronic inflammation by promoting Treg cell polarization and inhibiting the development of osteoclastogenic Th17 cells. Moreover, SP could rejuvenate stem cell and enable stem cells to repopulate and differentiate into osteoblast. Collectively, our study strongly suggests that SP treatment can block osteoporosis and furthermore, SP treatment provides therapeutic effect on chronic disease with inflammation and stem cell dysfunction.
Collapse
Affiliation(s)
- Jiyuan Piao
- Graduate School of Biotechnology and Department of Genetic Engineering, College of Life Science, Kyung Hee University, Seochun-dong, Kiheung-ku, Yong In, Republic of Korea
| | - Jeong Seop Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, Republic of Korea
| | - Dae Yeon Hwang
- Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| | - Youngsook Son
- Graduate School of Biotechnology and Department of Genetic Engineering, College of Life Science, Kyung Hee University, Seochun-dong, Kiheung-ku, Yong In, Republic of Korea.,Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, Republic of Korea.,Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea.,East-West Medical Research Institute, Kyung Hee University Hospital, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| |
Collapse
|
23
|
Haque N, Fareez IM, Fong LF, Mandal C, Kasim NHA, Kacharaju KR, Soesilawati P. Role of the CXCR4-SDF1-HMGB1 pathway in the directional migration of cells and regeneration of affected organs. World J Stem Cells 2020. [DOI: 10.4252/wjsc.v12.i9.0000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
24
|
Haque N, Fareez IM, Fong LF, Mandal C, Abu Kasim NH, Kacharaju KR, Soesilawati P. Role of the CXCR4-SDF1-HMGB1 pathway in the directional migration of cells and regeneration of affected organs. World J Stem Cells 2020; 12:938-951. [PMID: 33033556 PMCID: PMC7524697 DOI: 10.4252/wjsc.v12.i9.938] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/18/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
In recent years, several studies have reported positive outcomes of cell-based therapies despite insufficient engraftment of transplanted cells. These findings have created a huge interest in the regenerative potential of paracrine factors released from transplanted stem or progenitor cells. Interestingly, this notion has also led scientists to question the role of proteins in the secretome produced by cells, tissues or organisms under certain conditions or at a particular time of regenerative therapy. Further studies have revealed that the secretomes derived from different cell types contain paracrine factors that could help to prevent apoptosis and induce proliferation of cells residing within the tissues of affected organs. This could also facilitate the migration of immune, progenitor and stem cells within the body to the site of inflammation. Of these different paracrine factors present within the secretome, researchers have given proper consideration to stromal cell-derived factor-1 (SDF1) that plays a vital role in tissue-specific migration of the cells needed for regeneration. Recently researchers recognized that SDF1 could facilitate site-specific migration of cells by regulating SDF1-CXCR4 and/or HMGB1-SDF1-CXCR4 pathways which is vital for tissue regeneration. Hence in this study, we have attempted to describe the role of different types of cells within the body in facilitating regeneration while emphasizing the HMGB1-SDF1-CXCR4 pathway that orchestrates the migration of cells to the site where regeneration is needed.
Collapse
Affiliation(s)
- Nazmul Haque
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor 42610, Malaysia
| | - Ismail M Fareez
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor 42610, Malaysia
| | - Liew Fong Fong
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor 42610, Malaysia
| | - Chanchal Mandal
- Biotechnology and Genetic Engineering Discipline, Life Science, Khulna University, Khulna 9208, Bangladesh
| | - Noor Hayaty Abu Kasim
- Faculty of Dentistry, University Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
- Faculty of Dental Medicine, Universitas Airlangga, Surabaya 411007, Indonesia
| | - Kranthi Raja Kacharaju
- Department of Conservative Dentistry, Faculty of Dentistry MAHSA University, Selangor 42610, Malaysia
| | - Pratiwi Soesilawati
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| |
Collapse
|
25
|
Regulation of CXCR6 Expression on Adipocytes and Osteoblasts Differentiated from Human Adipose Tissue-Derived Mesenchymal Stem Cells. Stem Cells Int 2020; 2020:8870133. [PMID: 32922452 PMCID: PMC7453243 DOI: 10.1155/2020/8870133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 01/16/2023] Open
Abstract
Human mesenchymal stem cells derived from adipose tissue (hADMSCs) are a desirable candidate in regenerative medicine. hADMSCs secrete growth factors, cytokines, and chemokines and also express various receptors that are important in cell activation, differentiation, and migration to injured tissue. We showed that the expression level of chemokine receptor CXCR6 was significantly increased by ~2.5-fold in adipogenic-differentiated cells (Ad), but not in osteogenic-differentiated cells (Os) when compared with hADMSCs. However, regulation of CXCR6 expression on hADMSCs by using lentiviral particles did not affect the differentiation potential of hADMSCs. Increased expression of CXCR6 on Ad was mediated by both receptor recycling, which was in turn regulated by secretion of CXCL16, and de novo synthesis. The level of soluble CXCL16 was highly increased in both Ad and Os in particular, which inversely correlates with the expression on a transmembrane-bound form of CXCL16 that is cleaved by disintegrin and metalloproteinase. We concluded that the expression of CXCR6 is regulated by receptor degradation or recycling when it is internalized by interaction with CXCL16 and by de novo synthesis of CXCR6. Overall, our study may provide an insight into the molecular mechanisms of the CXCR6 reciprocally expressed on differentiated cells from hADMSCs.
Collapse
|
26
|
Zhang Y, Babczyk P, Pansky A, Kassack MU, Tobiasch E. P2 Receptors Influence hMSCs Differentiation towards Endothelial Cell and Smooth Muscle Cell Lineages. Int J Mol Sci 2020; 21:E6210. [PMID: 32867347 PMCID: PMC7503934 DOI: 10.3390/ijms21176210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Human mesenchymal stem cells (hMSCs) have shown their multipotential including differentiating towards endothelial and smooth muscle cell lineages, which triggers a new interest for using hMSCs as a putative source for cardiovascular regenerative medicine. Our recent publication has shown for the first time that purinergic 2 receptors are key players during hMSC differentiation towards adipocytes and osteoblasts. Purinergic 2 receptors play an important role in cardiovascular function when they bind to extracellular nucleotides. In this study, the possible functional role of purinergic 2 receptors during MSC endothelial and smooth muscle differentiation was investigated. METHODS AND RESULTS Human MSCs were isolated from liposuction materials. Then, endothelial and smooth muscle-like cells were differentiated and characterized by specific markers via Reverse Transcriptase-PCR (RT-PCR), Western blot and immunochemical stainings. Interestingly, some purinergic 2 receptor subtypes were found to be differently regulated during these specific lineage commitments: P2Y4 and P2Y14 were involved in the early stage commitment while P2Y1 was the key player in controlling MSC differentiation towards either endothelial or smooth muscle cells. The administration of natural and artificial purinergic 2 receptor agonists and antagonists had a direct influence on these differentiations. Moreover, a feedback loop via exogenous extracellular nucleotides on these particular differentiations was shown by apyrase digest. CONCLUSIONS Purinergic 2 receptors play a crucial role during the differentiation towards endothelial and smooth muscle cell lineages. Some highly selective and potent artificial purinergic 2 ligands can control hMSC differentiation, which might improve the use of adult stem cells in cardiovascular tissue engineering in the future.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany; (Y.Z.); (P.B.); (A.P.)
- Institute of Pharmaceutical & Medicinal Chemistry, University of Dusseldorf, D-40225 Dusseldorf, Germany;
| | - Patrick Babczyk
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany; (Y.Z.); (P.B.); (A.P.)
| | - Andreas Pansky
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany; (Y.Z.); (P.B.); (A.P.)
| | - Matthias Ulrich Kassack
- Institute of Pharmaceutical & Medicinal Chemistry, University of Dusseldorf, D-40225 Dusseldorf, Germany;
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany; (Y.Z.); (P.B.); (A.P.)
| |
Collapse
|
27
|
Saberian M, Shahidi Delshad E, Habibi M. The Effect of Bifidobacterium Bifidum Supernatant and Cell Mass on the Proliferation Potential of Rat Bone Marrow-Derived Stromal Cells. IRANIAN JOURNAL OF MEDICAL SCIENCES 2020; 45:269-276. [PMID: 32801416 PMCID: PMC7395953 DOI: 10.30476/ijms.2019.45772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background Mesenchymal stem cells (MSCs) are widely used to treat various diseases, however, their proliferative potential reduces after a number of passages. It has been shown that some probiotics such as Bifidobacterium Bifidum (B. bifidum) affect the proliferation of various cell lineages. The present study aimed to investigate the effect of B. bifidum on the proliferation of rat bone marrow stromal cells (rBMSCs) and to develop a method for compensating their proliferation reduction after some passages. Methods The present experimental study was conducted at Tehran University of Medical Sciences, Tehran, Iran, in 2017. The stromal cells were isolated from rBMSCs and their mesenchymal properties were confirmed by osteogenic and adipogenic differentiation media and staining. B. bifidum was cultured and the B. bifidum supernatant (BS) and bacterial cell mass (BCM) were extracted. The rBMSCs were treated with different concentrations of BS and BCM. The MTT assay was performed to measure the number of viable cells in the culture. Cell proliferation was analyzed using the paired-sample t test. Results Cell proliferation increased as the concentration of bacteria was increased logarithmically (0, 0.1, 0.3, 0.9, 3, 9, 30 μL/mL). In comparison with BS, cells treated with BCM showed increased cell proliferation at lower concentrations. This effect was caused by removing the "de Man, Rogosa, and Sharpe" (MRS) broth medium from the BCM culture. The optimal concentration of bacteria with the most significant effect on rBMSCs proliferation was determined. Conclusion A significant increase in the proliferation of stromal cells was observed; confirming the stimulatory potential of probiotics (B. bifidum) on various cells. The use of products containing probiotic bacteria can increase the proliferation potential of BMSCs.
Collapse
Affiliation(s)
- Mostafa Saberian
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences. Tehran, Iran
| | - Elham Shahidi Delshad
- Shahid Rajaei Cardiovascular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Habibi
- Department of Genetics, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
28
|
Turnbull G, Clarke J, Picard F, Zhang W, Riches P, Li B, Shu W. 3D biofabrication for soft tissue and cartilage engineering. Med Eng Phys 2020; 82:13-39. [PMID: 32709263 DOI: 10.1016/j.medengphy.2020.06.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 05/25/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023]
Abstract
Soft tissue injuries (STIs) affect patients of all age groups and represent a common worldwide clinical problem, resulting from conditions including trauma, infection, cancer and burns. Within the spectrum of STIs a mixture of tissues can be injured, ranging from skin to underlying nerves, blood vessels, tendons and cartilaginous tissues. However, significant limitations affect current treatment options and clinical demand for soft tissue and cartilage regenerative therapies continues to rise. Improving the regeneration of soft tissues has therefore become a key area of focus within tissue engineering. As an emerging technology, 3D bioprinting can be used to build complex soft tissue constructs "from the bottom up," by depositing cells, growth factors, extracellular matrices and other biomaterials in a layer-by-layer fashion. In this way, regeneration of cartilage, skin, vasculature, nerves, tendons and other bodily tissues can be performed in a patient specific manner. This review will focus on recent use of 3D bioprinting and other biofabrication strategies in soft tissue repair and regeneration. Biofabrication of a variety of soft tissue types will be reviewed following an overview of available cell sources, bioinks and bioprinting techniques.
Collapse
Affiliation(s)
- Gareth Turnbull
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow G4 0NW, United Kingdom; Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank G81 4DY, United Kingdom
| | - Jon Clarke
- Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank G81 4DY, United Kingdom
| | - Frédéric Picard
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow G4 0NW, United Kingdom; Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank G81 4DY, United Kingdom
| | - Weidong Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Philip Riches
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow G4 0NW, United Kingdom
| | - Bin Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Wenmiao Shu
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow G4 0NW, United Kingdom.
| |
Collapse
|
29
|
Woods DC, Wereszczynski J. Elucidating the influence of linker histone variants on chromatosome dynamics and energetics. Nucleic Acids Res 2020; 48:3591-3604. [PMID: 32128577 PMCID: PMC7144933 DOI: 10.1093/nar/gkaa121] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/23/2022] Open
Abstract
Linker histones are epigenetic regulators that bind to nucleosomes and alter chromatin structures and dynamics. Biophysical studies have revealed two binding modes in the linker histone/nucleosome complex, the chromatosome, where the linker histone is either centered on or askew from the dyad axis. Each has been posited to have distinct effects on chromatin, however the molecular and thermodynamic mechanisms that drive them and their dependence on linker histone compositions remain poorly understood. We present molecular dynamics simulations of chromatosomes with the globular domain of two linker histone variants, generic H1 (genGH1) and H1.0 (GH1.0), to determine how their differences influence chromatosome structures, energetics and dynamics. Results show that both unbound linker histones adopt a single compact conformation. Upon binding, DNA flexibility is reduced, resulting in increased chromatosome compaction. While both variants enthalpically favor on-dyad binding, energetic benefits are significantly higher for GH1.0, suggesting that GH1.0 is more capable than genGH1 of overcoming the large entropic reduction required for on-dyad binding which helps rationalize experiments that have consistently demonstrated GH1.0 in on-dyad states but that show genGH1 in both locations. These simulations highlight the thermodynamic basis for different linker histone binding motifs, and details their physical and chemical effects on chromatosomes.
Collapse
Affiliation(s)
- Dustin C Woods
- Department of Chemistry and the Center for Molecular Study of Condensed Soft Matter, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Jeff Wereszczynski
- Department of Physics and the Center for Molecular Study of Condensed Soft Matter, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
30
|
Periyasamy-Thandavan S, Burke J, Mendhe B, Kondrikova G, Kolhe R, Hunter M, Isales CM, Hamrick MW, Hill WD, Fulzele S. MicroRNA-141-3p Negatively Modulates SDF-1 Expression in Age-Dependent Pathophysiology of Human and Murine Bone Marrow Stromal Cells. J Gerontol A Biol Sci Med Sci 2020; 74:1368-1374. [PMID: 31505568 DOI: 10.1093/gerona/gly186] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Indexed: 12/11/2022] Open
Abstract
Stromal cell-derived factor-1 (SDF-1 or CXCL12) is a cytokine secreted by cells including bone marrow stromal cells (BMSCs). SDF-1 plays a vital role in BMSC migration, survival, and differentiation. Our group previously reported the role of SDF-1 in osteogenic differentiation in vitro and bone formation in vivo; however, our understanding of the post-transcriptional regulatory mechanism of SDF-1 remains poor. MicroRNAs are small noncoding RNAs that post-transcriptionally regulate the messenger RNAs (mRNAs) of protein-coding genes. In this study, we aimed to investigate the impact of miR-141-3p on SDF-1 expression in BMSCs and its importance in the aging bone marrow (BM) microenvironment. Our data demonstrated that murine and human BMSCs expressed miR-141-3p that repressed SDF-1 gene expression at the functional level (luciferase reporter assay) by targeting the 3'-untranslated region of mRNA. We also found that transfection of miR-141-3p decreased osteogenic markers in human BMSCs. Our results demonstrate that miR-141-3p expression increases with age, while SDF-1 decreases in both the human and mouse BM niche. Taken together, these results support that miR-141-3p is a novel regulator of SDF-1 in bone cells and plays an important role in the age-dependent pathophysiology of murine and human BM niche.
Collapse
Affiliation(s)
| | - John Burke
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia
| | - Bharati Mendhe
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia
| | - Galina Kondrikova
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, Georgia
| | - Monte Hunter
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia
| | - Carlos M Isales
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Georgia
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia.,Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Georgia
| | - William D Hill
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia.,Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Georgia.,Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Sadanand Fulzele
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia.,Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Georgia
| |
Collapse
|
31
|
Jacques C, Tesfaye R, Lavaud M, Georges S, Baud’huin M, Lamoureux F, Ory B. Implication of the p53-Related miR-34c, -125b, and -203 in the Osteoblastic Differentiation and the Malignant Transformation of Bone Sarcomas. Cells 2020; 9:cells9040810. [PMID: 32230926 PMCID: PMC7226610 DOI: 10.3390/cells9040810] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
Abstract
The formation of the skeleton occurs throughout the lives of vertebrates and is achieved through the balanced activities of two kinds of specialized bone cells: the bone-forming osteoblasts and the bone-resorbing osteoclasts. Impairment in the remodeling processes dramatically hampers the proper healing of fractures and can also result in malignant bone diseases such as osteosarcoma. MicroRNAs (miRNAs) are a class of small non-coding single-strand RNAs implicated in the control of various cellular activities such as proliferation, differentiation, and apoptosis. Their post-transcriptional regulatory role confers on them inhibitory functions toward specific target mRNAs. As miRNAs are involved in the differentiation program of precursor cells, it is now well established that this class of molecules also influences bone formation by affecting osteoblastic differentiation and the fate of osteoblasts. In response to various cell signals, the tumor-suppressor protein p53 activates a huge range of genes, whose miRNAs promote genomic-integrity maintenance, cell-cycle arrest, cell senescence, and apoptosis. Here, we review the role of three p53-related miRNAs, miR-34c, -125b, and -203, in the bone-remodeling context and, in particular, in osteoblastic differentiation. The second aim of this study is to deal with the potential implication of these miRNAs in osteosarcoma development and progression.
Collapse
|
32
|
He W, Zheng Y, Feng Q, Elkhooly TA, Liu X, Yang X, Wang Y, Xie Y. Silver nanoparticles stimulate osteogenesis of human mesenchymal stem cells through activation of autophagy. Nanomedicine (Lond) 2020; 15:337-353. [DOI: 10.2217/nnm-2019-0026] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: Previously, different results have been achieved regarding effects of silver nanoparticles (Ag NPs) on osteogenesis of stem cells and the mechanisms have not been disclosed yet, which are quite important for potential application of Ag NPs in bone reconstruction. Materials & methods: Effects of Ag NPs on osteogenesis of human mesenchymal stem cells (hMSCs) with underlying mechanisms were investigated. Results: Ag NPs at 2.5 and 5 μg/ml increased osteogenic proteins expression and mineralization of hMSCs. Meanwhile, autophagy was activated by Ag NPs and it could be inhibited by 3-methyladenine. Furthermore, osteogenesis induced by Ag NPs could also be reversed by 3-methyladenine. Conclusion: These findings suggest that autophagy is involved in stimulating osteogenesis of hMSCs induced by Ag NPs.
Collapse
Affiliation(s)
- Wei He
- School of Materials Science & Engineering, University of Science & Technology Beijing, Beijing, 100083, PR China
| | - Yudong Zheng
- School of Materials Science & Engineering, University of Science & Technology Beijing, Beijing, 100083, PR China
| | - Qingling Feng
- State Key Laboratory of New Ceramics & Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Tarek A Elkhooly
- Department of Refractories, Ceramics & Building Materials, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Xujie Liu
- State Key Laboratory of New Ceramics & Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Xing Yang
- State Key Laboratory of New Ceramics & Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Yansen Wang
- School of Materials Science & Engineering, University of Science & Technology Beijing, Beijing, 100083, PR China
| | - Yajie Xie
- School of Materials Science & Engineering, University of Science & Technology Beijing, Beijing, 100083, PR China
| |
Collapse
|
33
|
Melatonin Reverses the Loss of Stemness Induced by TNF- α in Human Bone Marrow Mesenchymal Stem Cells through Upregulation of YAP Expression. Stem Cells Int 2019; 2019:6568394. [PMID: 32082385 PMCID: PMC7012241 DOI: 10.1155/2019/6568394] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising candidates for tissue regeneration and disease treatment. However, long-term in vitro culture results in loss of MSC stemness. The inflammation that occurs at stem cell transplant sites (such as that resulting from TNF-α) is a contributing factor for stem cell treatment failure. Currently, there is little evidence regarding the protective role of melatonin with regard to the negative effects of TNF-α on the stemness of MSCs. In this study, we report a melatonin-based method to reduce the inflammatory effects on the stemness of bone marrow mesenchymal stem cells (BMMSCs). The results of colony formation assays, Alizarin red staining, western blotting, and reverse transcription-polymerase chain reactions suggest that melatonin can reverse the inflammatory damage caused by TNF-α treatment in the third, seventh, and tenth generations of primary BMMSCs (vs. control and the TNF-α-treated group). Meanwhile, a detailed analysis of the molecular mechanisms showed that the melatonin receptor and YAP signaling pathway are closely related to the role that melatonin plays in negative inflammatory effects against BMMSCs. In addition, in vivo experiments showed that melatonin could reverse the damage caused by TNF-α on bone regeneration by BMMSCs in nude mice. Overall, our results suggest that melatonin can reverse the loss of stemness caused by inflammatory factor TNF-α in BMMSCs. Our results also provide a practical strategy for the application of BMMSCs in tissue engineering and cell therapy.
Collapse
|
34
|
Shao X, Ai G, Wang L, Qin J, Li Y, Jiang H, Zhang T, Zhou L, Gao Z, Cheng J, Cheng Z. Adipose-derived stem cells transplantation improves endometrial injury repair. ZYGOTE 2019; 27:367-374. [PMID: 31452481 DOI: 10.1017/s096719941900042x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endometrial injury is an important cause of intrauterine adhesion (IUA), amenorrhea and infertility in women, with limited effective therapies. Recently, stem cells have been used in animal experiments to repair and improve injured endometrium. To date, our understanding of adipose-derived stem cells (ADSCs) in endometrial injury repair and their further therapeutic mechanisms is incomplete. Here, we examined the benefit of ADSCs in restoration of injured endometrium by applying a rat endometrial injury model. The results revealed by immunofluorescence showed that green fluorescent protein (GFP)-labelled ADSCs can differentiate into endometrial epithelial cells in vivo. At 30 days after ADSCs transplantation, injured endometrium was significantly improved, with increased microvessel density, endometrial thickness and glands when compared with the model group. Furthermore, the fertility of rats with injured endometrium in ADSCs group was improved and had a higher conception rate (60% vs 20%, P = 0.014) compared with the control phosphate-buffered saline (PBS) group. However, there was no difference in the control group compared with the sham group. In addition, expression levels of the oestrogen receptor Eα/β (ERα, ERβ) and progesterone receptor (PR) detected by western blot and enzyme-linked immunosorbent assay (ELISA) were higher in the ADSCs group than in the PBS group. Taken together, these results suggested that ADSC transplantation could improve endometrial injury as a novel therapy for IUA.
Collapse
Affiliation(s)
- Xiaowen Shao
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Guihai Ai
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Lian Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jinlong Qin
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yue Li
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Huici Jiang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Tingting Zhang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Linlin Zhou
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhengliang Gao
- Lifeng Institute of Regenerative Medicine, Tongji University, Shanghai, 200092, China
| | - Jiajing Cheng
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhongping Cheng
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| |
Collapse
|
35
|
Development and Evaluation of a Prototype Scratch Apparatus for Wound Assays Adjustable to Different Forces and Substrates. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9204414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Scratch assays enable the study of the migration process of an injured adherent cell layer in vitro. An apparatus for the reproducible performance of scratch assays and cell harvesting has been developed that meets the requirements for reproducibility in tests as well as easy handling. The entirely autoclavable setup is divided into a sample translation and a scratching system. The translational system is compatible with standard culture dishes and can be modified to adapt to different cell culture systems, while the scratching system can be adjusted according to angle, normal force, shape, and material to adapt to specific questions and demanding substrates. As a result, a fully functional prototype can be presented. This system enables the creation of reproducible and clear scratch edges with a low scratch border roughness within a monolayer of cells. Moreover, the apparatus allows the collection of the migrated cells after scratching for further molecular biological investigations without the need for a second processing step. For comparison, the mechanical properties of manually performed scratch assays are evaluated.
Collapse
|
36
|
Qi Y, Ma J, Li S, Liu W. Applicability of adipose-derived mesenchymal stem cells in treatment of patients with type 2 diabetes. Stem Cell Res Ther 2019; 10:274. [PMID: 31455405 PMCID: PMC6712852 DOI: 10.1186/s13287-019-1362-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is mainly characterized by insulin resistance (IR) and impaired insulin secretion. The chronic inflammatory process contributed to IR and could also hamper pancreatic β cell function. However, currently applied treatment cannot reverse β cell damage or alleviate inflammation. Mesenchymal stem cells (MSCs), the cell-based therapy for their self-renewable, differentiation potential, and immunosuppressive properties, have been demonstrated in displaying therapeutic effects in T2DM. Adipose-derived MSCs (AD-MSCs) attracted more attention due to less harvested inconvenience and ethical issues commonly accompany with bone marrow-derived MSCs (BM-MSCs) and fetal annex-derived MSCs. Both AD-MSC therapy studies and mechanism explorations in T2DM animals presented that AD-MSCs could translate to clinical application. However, hyperglycemia, hyperinsulinemia, and metabolic disturbance in T2DM are crucial for impairment of AD-MSC function, which may limit the therapeutical effects of MSCs. This review focuses on the outcomes and the molecular mechanisms of MSC therapies in T2DM which light up the hope of AD-MSCs as an innovative strategy to cure T2DM.
Collapse
Affiliation(s)
- Yicheng Qi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, School of Medicine, Shanghai Jiaotong University, 160# Pujian Road, Pudong, Shanghai, 200127, China
| | - Jing Ma
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, School of Medicine, Shanghai Jiaotong University, 160# Pujian Road, Pudong, Shanghai, 200127, China
| | - Shengxian Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, School of Medicine, Shanghai Jiaotong University, 160# Pujian Road, Pudong, Shanghai, 200127, China
| | - Wei Liu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, School of Medicine, Shanghai Jiaotong University, 160# Pujian Road, Pudong, Shanghai, 200127, China.
| |
Collapse
|
37
|
Pierce JL, Ding KH, Xu J, Sharma AK, Yu K, Del Mazo Arbona N, Rodriguez-Santos Z, Bernard P, Bollag WB, Johnson MH, Hamrick MW, Begun DL, Shi XM, Isales CM, McGee-Lawrence ME. The glucocorticoid receptor in osteoprogenitors regulates bone mass and marrow fat. J Endocrinol 2019; 243:JOE-19-0230.R1. [PMID: 31370004 PMCID: PMC6938567 DOI: 10.1530/joe-19-0230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022]
Abstract
Excess fat within bone marrow is associated with lower bone density. Metabolic stressors such as chronic caloric restriction (CR) can exacerbate marrow adiposity, and increased glucocorticoid signaling and adrenergic signaling are implicated in this phenotype. The current study tested the role of glucocorticoid signaling in CR-induced stress by conditionally deleting the glucocorticoid receptor (GR) in bone marrow osteoprogenitors (Osx1-Cre) of mice subjected to CR and ad libitum diets. Conditional knockout of the GR (GR-CKO) reduced cortical and trabecular bone mass as compared to wildtype (WT) mice under both ad libitum and CR conditions. No interaction was detected between genotype and diet, suggesting that the GR is not required for CR-induced skeletal changes. The lower bone mass in GR-CKO mice, and the further suppression of bone by CR, resulted from suppressed bone formation. Interestingly, treatment with the -adrenergic receptor antagonist propranolol mildly but selectively improved metrics of cortical bone mass in GR-CKO mice during CR, suggesting interaction between adrenergic and glucocorticoid signaling pathways that affects cortical bone. GR-CKO mice dramatically increased marrow fat under both ad libitum and CR-fed conditions, and surprisingly propranolol treatment was unable to rescue CR-induced marrow fat in either WT or GR-CKO mice. Additionally, serum corticosterone levels were selectively elevated in GR-CKO mice with CR, suggesting the possibility of bone-hypothalamus-pituitary-adrenal crosstalk during metabolic stress. This work highlights the complexities of glucocorticoid and β-adrenergic signaling in stress-induced changes in bone mass, and the importance of GR function in suppressing marrow adipogenesis while maintaining healthy bone mass.
Collapse
Affiliation(s)
- Jessica L Pierce
- J Pierce, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | - Ke-Hong Ding
- K Ding, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Jianrui Xu
- J Xu, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Anuj K Sharma
- A Sharma, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | - Kanglun Yu
- K Yu, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | | | | | - Paul Bernard
- P Bernard, Pediatric Endocrine Specialists of Georgia, Pediatric Endocrine Specialists of Georgia, Duluth, United States
| | - Wendy B Bollag
- W Bollag, Department of Physiology, Medical College of Georgia, Augusta, GA 30912, United States
| | - Maribeth H Johnson
- M Johnson, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Mark W Hamrick
- M Hamrick, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | - Dana L Begun
- D Begun, Department of Orthopedic Surgery, Mayo Clinic, Rochester, United States
| | - Xing M Shi
- X Ming Shi, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Carlos M Isales
- C Isales, Neuroscience and Regenerative Medicine, Augusta University, Augusta, 30912, United States
| | | |
Collapse
|
38
|
Żurek A, Mizerska-Kowalska M, Sławińska-Brych A, Kaławaj K, Bojarska-Junak A, Kandefer-Szerszeń M, Zdzisińska B. Alpha ketoglutarate exerts a pro-osteogenic effect in osteoblast cell lines through activation of JNK and mTOR/S6K1/S6 signaling pathways. Toxicol Appl Pharmacol 2019; 374:53-64. [PMID: 31051157 DOI: 10.1016/j.taap.2019.04.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/30/2019] [Accepted: 04/27/2019] [Indexed: 01/18/2023]
Abstract
Although numerous in vivo studies have suggested that alpha-ketoglutarate (AKG), i.e. the key intermediate in the Krebs cycle, may have an anabolic effect on bone tissue, the direct influence of AKG on osteoblasts and the underlying mechanism of its action have not been investigated so far. The aim of this study was to assess the impact of AKG (disodium salt dihydrate) on osteogenesis in vitro and identification of some signaling mechanisms involved in this activity. The human and mouse normal osteoblast cell lines hFOB 1.19 and MC3T3-E1 were used in this study. The results showed that AKG did not increase the proliferation of osteoblasts; however, it upregulated the expression of transcription factors RUNX2 and Osterix, the mRNA and protein levels of osteoblast differentiation markers (alkaline phosphatase, type I collagen, bone sialoprotein II, osteopontin, osteocalcin), and the mineralization levels in the hFOB 1.19 and MC3T3-E1 cell cultures. Moreover, AKG increased JNK, mTOR, S6K1, and S6 phosphorylation and decreased ERK1/2 phosphorylation in both osteoblast cell lines. The JNK inhibitor and rapamycin, but not the ERK inhibitor, abolished the AKG-promoted osteoblast differentiation. Using immunofluorescence staining, qRT-PCR, and Western blot analysis, we detected the presence of an AKG receptor GPR99 activated by alpha ketoglutaric acid in the tested osteoblast cell lines. However, AKG salt did not activate GPR99. Our findings suggest that AKG salt activates the JNK and mTOR/S6K1/S6 signaling pathways to promote differentiation of osteoblasts, independently of GPR99 activation. We can conclude that AKG salts might be promising candidates for bone anabolic drugs used for prevention or/and treatment of osteoporosis.
Collapse
Affiliation(s)
- Aleksandra Żurek
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Lublin, Poland
| | | | | | - Katarzyna Kaławaj
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Lublin, Poland
| | | | | | - Barbara Zdzisińska
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Lublin, Poland.
| |
Collapse
|
39
|
Li Y, Liao C, Tjong SC. Synthetic Biodegradable Aliphatic Polyester Nanocomposites Reinforced with Nanohydroxyapatite and/or Graphene Oxide for Bone Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E590. [PMID: 30974820 PMCID: PMC6523566 DOI: 10.3390/nano9040590] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/22/2019] [Accepted: 04/03/2019] [Indexed: 12/27/2022]
Abstract
This paper provides review updates on the current development of bionanocomposites with polymeric matrices consisting of synthetic biodegradable aliphatic polyesters reinforced with nanohydroxyaptite (nHA) and/or graphene oxide (GO) nanofillers for bone tissue engineering applications. Biodegradable aliphatic polyesters include poly(lactic acid) (PLA), polycaprolactone (PCL) and copolymers of PLA-PGA (PLGA). Those bionanocomposites have been explored for making 3D porous scaffolds for the repair of bone defects since nHA and GO enhance their bioactivity and biocompatibility by promoting biomineralization, bone cell adhesion, proliferation and differentiation, thus facilitating new bone tissue formation upon implantation. The incorporation of nHA or GO into aliphatic polyester scaffolds also improves their mechanical strength greatly, especially hybrid GO/nHA nanofilllers. Those mechanically strong nanocomposite scaffolds can support and promote cell attachment for tissue growth. Porous scaffolds fabricated from conventional porogen leaching, and thermally induced phase separation have many drawbacks inducing the use of organic solvents, poor control of pore shape and pore interconnectivity, while electrospinning mats exhibit small pores that limit cell infiltration and tissue ingrowth. Recent advancement of 3D additive manufacturing allows the production of aliphatic polyester nanocomposite scaffolds with precisely controlled pore geometries and large pores for the cell attachment, growth, and differentiation in vitro, and the new bone formation in vivo.
Collapse
Affiliation(s)
- Yuchao Li
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng 252059, China.
| | - Chengzhu Liao
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Sie Chin Tjong
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China.
| |
Collapse
|
40
|
Effects of different intensities of strength and endurance training on some osteometabolic miRNAs, Runx2 and PPARγ in bone marrow of old male wistar rats. Mol Biol Rep 2019; 46:2513-2521. [PMID: 30915688 DOI: 10.1007/s11033-019-04695-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 02/09/2019] [Indexed: 10/27/2022]
Abstract
Bone tissue is known as a living dynamic and complex organ in response to physical activity and mechanical loading such as exercise training; thus, the purpose of this study was to determine the effect of different intensities of strength and endurance training on expression of some osteometabolic miRNAs and runt-related transcription factor 2 (Runx2) and peroxisome proliferator-activated receptor γ (PPARγ) in bone marrow of old male Wistar rats. To this end, a total number of 50 male Wistar rats (aged 23 months, 438.27 g) were obtained from Pasteur Institute of Iran. The rats were randomized into five groups (10 rats/per group) including moderate endurance training (MET), high-intensity endurance training (HET), moderate-intensity resistance training (MRT), high-intensity resistance training (HRT), and control (CON). The four training groups completed 8 weeks of a training program, 5 days a week, according to the study protocol. To evaluate miR-133a, miR-103a, miR-204, and other adipogenic and osteogenic genes such as RUNX2 and PPARγ via real-time PCR, total RNA including mRNA and miRNA was isolated from the bone marrow. The statistical analysis was then performed using two-way analysis of variance (ANOVA). No significant differences in miR-133a (p = 0.197), miR-103a (p = 0.302), miR-204 (p = 0.539), RUNX2 (p = 0.960), and PPARγ (P = 0.872) were observed between the intervention groups and the control one. Furthermore, there were no significant differences in bone force (p = 0.641), fracture energy (p = 0.982), stress (p = 0.753), module (p = 0.147), and elongation (p = 0.292) variables between the intervention groups and the control group. Investigating molecular and cellular changes in the bone after such exercises in longer time could provide clearer results about the beneficial or harmful effects of these types of exercises in healthy and passive elderly people.
Collapse
|
41
|
Götz W, Tobiasch E, Witzleben S, Schulze M. Effects of Silicon Compounds on Biomineralization, Osteogenesis, and Hard Tissue Formation. Pharmaceutics 2019; 11:E117. [PMID: 30871062 PMCID: PMC6471146 DOI: 10.3390/pharmaceutics11030117] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/25/2019] [Accepted: 03/03/2019] [Indexed: 12/19/2022] Open
Abstract
Bioinspired stem cell-based hard tissue engineering includes numerous aspects: The synthesis and fabrication of appropriate scaffold materials, their analytical characterization, and guided osteogenesis using the sustained release of osteoinducing and/or osteoconducting drugs for mesenchymal stem cell differentiation, growth, and proliferation. Here, the effect of silicon- and silicate-containing materials on osteogenesis at the molecular level has been a particular focus within the last decade. This review summarizes recently published scientific results, including material developments and analysis, with a special focus on silicon hybrid bone composites. First, the sources, bioavailability, and functions of silicon on various tissues are discussed. The second focus is on the effects of calcium-silicate biomineralization and corresponding analytical methods in investigating osteogenesis and bone formation. Finally, recent developments in the manufacturing of Si-containing scaffolds are discussed, including in vitro and in vivo studies, as well as recently filed patents that focus on the influence of silicon on hard tissue formation.
Collapse
Affiliation(s)
- Werner Götz
- Department of Orthodontics, Oral Biology Laboratory, School of Dentistry, Rheinische Wilhelms University of Bonn, Welschnonnenstr. 17, D-53111 Bonn, Germany.
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Steffen Witzleben
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| |
Collapse
|
42
|
Pierce JL, Begun DL, Westendorf JJ, McGee-Lawrence ME. Defining osteoblast and adipocyte lineages in the bone marrow. Bone 2019; 118:2-7. [PMID: 29782940 PMCID: PMC6240509 DOI: 10.1016/j.bone.2018.05.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/16/2018] [Accepted: 05/16/2018] [Indexed: 12/22/2022]
Abstract
Bone is a complex endocrine organ that facilitates structural support, protection to vital organs, sites for hematopoiesis, and calcium homeostasis. The bone marrow microenvironment is a heterogeneous niche consisting of multipotent musculoskeletal and hematopoietic progenitors and their derivative terminal cell types. Amongst these progenitors, bone marrow mesenchymal stem/stromal cells (BMSCs) may differentiate into osteogenic, adipogenic, myogenic, and chondrogenic lineages to support musculoskeletal development as well as tissue homeostasis, regeneration and repair during adulthood. With age, the commitment of BMSCs to osteogenesis slows, bone formation decreases, fracture risk rises, and marrow adiposity increases. An unresolved question is whether osteogenesis and adipogenesis are co-regulated in the bone marrow. Osteogenesis and adipogenesis are controlled by specific signaling mechanisms, circulating cytokines, and transcription factors such as Runx2 and Pparγ, respectively. One hypothesis is that adipogenesis is the default pathway if osteogenic stimuli are absent. However, recent work revealed that Runx2 and Osx1-expressing preosteoblasts form lipid droplets under pathological and aging conditions. Histone deacetylase 3 (Hdac3) and other epigenetic regulators suppress lipid storage in preosteoblasts and/or control marrow adiposity. Establishing a better understanding of fat storage in bone marrow cells, as well as the osteoblast-adipocyte relationship within the bone marrow niche is necessary to understand the mechanisms underlying disease- and aging-related marrow fat storage and may lead to the development of new therapeutic targets for "fatty bone" and osteoporosis.
Collapse
Affiliation(s)
- J L Pierce
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - D L Begun
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - J J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - M E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA.
| |
Collapse
|
43
|
Ottensmeyer PF, Witzler M, Schulze M, Tobiasch E. Small Molecules Enhance Scaffold-Based Bone Grafts via Purinergic Receptor Signaling in Stem Cells. Int J Mol Sci 2018; 19:E3601. [PMID: 30441872 PMCID: PMC6274752 DOI: 10.3390/ijms19113601] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022] Open
Abstract
The need for bone grafts is high, due to age-related diseases, such as tumor resections, but also accidents, risky sports, and military conflicts. The gold standard for bone grafting is the use of autografts from the iliac crest, but the limited amount of accessible material demands new sources of bone replacement. The use of mesenchymal stem cells or their descendant cells, namely osteoblast, the bone-building cells and endothelial cells for angiogenesis, combined with artificial scaffolds, is a new approach. Mesenchymal stem cells (MSCs) can be obtained from the patient themselves, or from donors, as they barely cause an immune response in the recipient. However, MSCs never fully differentiate in vitro which might lead to unwanted effects in vivo. Interestingly, purinergic receptors can positively influence the differentiation of both osteoblasts and endothelial cells, using specific artificial ligands. An overview is given on purinergic receptor signaling in the most-needed cell types involved in bone metabolism-namely osteoblasts, osteoclasts, and endothelial cells. Furthermore, different types of scaffolds and their production methods will be elucidated. Finally, recent patents on scaffold materials, as wells as purinergic receptor-influencing molecules which might impact bone grafting, are discussed.
Collapse
Affiliation(s)
- Patrick Frank Ottensmeyer
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Markus Witzler
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| |
Collapse
|
44
|
Jiang H, Hong T, Wang T, Wang X, Cao L, Xu X, Zheng M. Gene expression profiling of human bone marrow mesenchymal stem cells during osteogenic differentiation. J Cell Physiol 2018; 234:7070-7077. [PMID: 30378112 DOI: 10.1002/jcp.27461] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/29/2018] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Osteogenesis is a multiple-step process through which osteoblasts are derived from bone marrow mesenchymal stem cells (MSCs) with multilineage differentiation potential. This study aimed to analyze gene expression profiling during osteogenic differentiation of MSCs. MATERIALS AND METHODS Human MSCs were isolated and induced for differentiation in osteogenic medium. Full-genome gene expression microarrays and gene ontology analysis were performed. RESULTS A total of 1,680 differentially expressed genes in differentiated MSCs were identified including 430 upregulated and 1,250 downregulated. Moreover, pathway-act-network analysis showed that cell cycle, p53 signaling pathway and focal adhesion, had high degree (>5). The ribonucleotide reductase M1, thymidine kinase 1 and histone cluster 1 H3e also showed high degree (>10). Polymerase chain reaction analysis confirmed the differential expression of insulin-like growth factor binding protein 3, SMAD family member 3, transforming growth factor beta 2, and fibroblast growth factor 14 in differentiated MSCs. CONCLUSIONS Gene expression profiling provides a foundation to reveal the mechanisms that regulate osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- He Jiang
- Key Laboratory for System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi, China
| | - Tao Hong
- Department of Ultrasound, The First Hospital of Jiujiang City, Jiangxi, China
| | - Tao Wang
- Key Laboratory for System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi, China
| | - Xinping Wang
- Key Laboratory for System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi, China
| | - Lingling Cao
- Department of Ultrasound, The First Hospital of Jiujiang City, Jiangxi, China
| | - Xiaoyuan Xu
- Key Laboratory for System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi, China
| | - Meirong Zheng
- Key Laboratory for System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
45
|
Witzler M, Alzagameem A, Bergs M, Khaldi-Hansen BE, Klein SE, Hielscher D, Kamm B, Kreyenschmidt J, Tobiasch E, Schulze M. Lignin-Derived Biomaterials for Drug Release and Tissue Engineering. Molecules 2018; 23:E1885. [PMID: 30060536 PMCID: PMC6222784 DOI: 10.3390/molecules23081885] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 12/29/2022] Open
Abstract
Renewable resources are gaining increasing interest as a source for environmentally benign biomaterials, such as drug encapsulation/release compounds, and scaffolds for tissue engineering in regenerative medicine. Being the second largest naturally abundant polymer, the interest in lignin valorization for biomedical utilization is rapidly growing. Depending on its resource and isolation procedure, lignin shows specific antioxidant and antimicrobial activity. Today, efforts in research and industry are directed toward lignin utilization as a renewable macromolecular building block for the preparation of polymeric drug encapsulation and scaffold materials. Within the last five years, remarkable progress has been made in isolation, functionalization and modification of lignin and lignin-derived compounds. However, the literature so far mainly focuses lignin-derived fuels, lubricants and resins. The purpose of this review is to summarize the current state of the art and to highlight the most important results in the field of lignin-based materials for potential use in biomedicine (reported in 2014⁻2018). Special focus is placed on lignin-derived nanomaterials for drug encapsulation and release as well as lignin hybrid materials used as scaffolds for guided bone regeneration in stem cell-based therapies.
Collapse
Affiliation(s)
- Markus Witzler
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, D-53359 Rheinbach, Germany.
| | - Abla Alzagameem
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, D-53359 Rheinbach, Germany.
- Faculty of Environment and Natural Sciences, Brandenburg University of Technology BTU Cottbus-Senftenberg, Platz der Deutschen Einheit 1, D-03046 Cottbus, Germany.
| | - Michel Bergs
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, D-53359 Rheinbach, Germany.
- Rheinische Friedrich-Wilhelms-University Bonn, INRES, Klein-Altendorf 2, D-53359 Rheinbach, Germany.
| | - Basma El Khaldi-Hansen
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, D-53359 Rheinbach, Germany.
| | - Stephanie E Klein
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, D-53359 Rheinbach, Germany.
| | - Dorothee Hielscher
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, D-53359 Rheinbach, Germany.
| | - Birgit Kamm
- Faculty of Environment and Natural Sciences, Brandenburg University of Technology BTU Cottbus-Senftenberg, Platz der Deutschen Einheit 1, D-03046 Cottbus, Germany.
- Kompetenzzentrum Holz GmbH, Altenberger Strasse 69, A-4040 Linz, Austria.
| | - Judith Kreyenschmidt
- Rheinische Friedrich Wilhelms-University Bonn, Katzenburgweg 7-9, D-53115 Bonn, Germany.
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, D-53359 Rheinbach, Germany.
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, D-53359 Rheinbach, Germany.
| |
Collapse
|
46
|
The role of OPG/RANKL in the pathogenesis of diabetic cardiovascular disease. Cardiovasc Endocrinol Metab 2018; 7:28-33. [PMID: 31646276 DOI: 10.1097/xce.0000000000000144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/21/2017] [Indexed: 11/25/2022]
Abstract
Cardiovascular (CV) disease is the leading cause of mortality in patients with type 2 diabetes mellitus. A major factor in the pathogenesis of CV disease is vascular calcification (VC), which is accelerated in type 2 diabetes mellitus. Calcification of the vessel wall contributes to vascular stiffness and left ventricular hypertrophy whereas intimal calcification may predispose to plaque rupture and CV death. The pathogenesis of VC is complex but appears to be regulated by the osteoprotegerin (OPG)/receptor activator of nuclear factor-κB ligand (RANKL) signaling pathway, which is involved in bone remodeling. Within the bone, OPG prevents RANKL from binding to receptor activator of nuclear factor-κB and inhibiting bone resorption. Outside of the bone, the clinical significance of OPG blocking RANKL is not well understood, but OPG knockout mice that lack OPG develop early and severe VC. This minireview outlines some of the research on OPG/RANKL in the pathogenesis of VC and discusses potential therapies, which may reduce VC and CV burden in humans.
Collapse
|
47
|
Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka P, Kasalkova NS, Svorcik V, Kolska Z, Motarjemi H, Molitor M. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells - a review. Biotechnol Adv 2018; 36:1111-1126. [PMID: 29563048 DOI: 10.1016/j.biotechadv.2018.03.011] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
Stem cells can be defined as units of biological organization that are responsible for the development and the regeneration of organ and tissue systems. They are able to renew their populations and to differentiate into multiple cell lineages. Therefore, these cells have great potential in advanced tissue engineering and cell therapies. When seeded on synthetic or nature-derived scaffolds in vitro, stem cells can be differentiated towards the desired phenotype by an appropriate composition, by an appropriate architecture, and by appropriate physicochemical and mechanical properties of the scaffolds, particularly if the scaffold properties are combined with a suitable composition of cell culture media, and with suitable mechanical, electrical or magnetic stimulation. For cell therapy, stem cells can be injected directly into damaged tissues and organs in vivo. Since the regenerative effect of stem cells is based mainly on the autocrine production of growth factors, immunomodulators and other bioactive molecules stored in extracellular vesicles, these structures can be isolated and used instead of cells for a novel therapeutic approach called "stem cell-based cell-free therapy". There are four main sources of stem cells, i.e. embryonic tissues, fetal tissues, adult tissues and differentiated somatic cells after they have been genetically reprogrammed, which are referred to as induced pluripotent stem cells (iPSCs). Although adult stem cells have lower potency than the other three stem cell types, i.e. they are capable of differentiating into only a limited quantity of specific cell types, these cells are able to overcome the ethical and legal issues accompanying the application of embryonic and fetal stem cells and the mutational effects associated with iPSCs. Moreover, adult stem cells can be used in autogenous form. These cells are present in practically all tissues in the organism. However, adipose tissue seems to be the most advantageous tissue from which to isolate them, because of its abundancy, its subcutaneous location, and the need for less invasive techniques. Adipose tissue-derived stem cells (ASCs) are therefore considered highly promising in present-day regenerative medicine.
Collapse
Affiliation(s)
- Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic.
| | - Jana Zarubova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Martina Travnickova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Julia Pajorova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Petr Slepicka
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Nikola Slepickova Kasalkova
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Vaclav Svorcik
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Zdenka Kolska
- Faculty of Science, J.E. Purkyne University, Ceske mladeze 8, 400 96 Usti nad Labem, Czech Republic
| | - Hooman Motarjemi
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| | - Martin Molitor
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| |
Collapse
|
48
|
Kruk JS, Bermeo S, Skarratt KK, Fuller SJ, Duque G. The Effect of Antidepressants on Mesenchymal Stem Cell Differentiation. J Bone Metab 2018; 25:43-51. [PMID: 29564305 PMCID: PMC5854822 DOI: 10.11005/jbm.2018.25.1.43] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 11/25/2022] Open
Abstract
Background Use of antidepressant medications has been linked to detrimental impacts on bone mineral density and osteoporosis; however, the cellular basis behind these observations remains poorly understood. The effect does not appear to be homogeneous across the whole class of drugs and may be linked to affinity for the serotonin transporter system. In this study, we hypothesized that antidepressants have a class- and dose-dependent effect on mesenchymal stem cell (MSC) differentiation, which may affect bone metabolism. Methods Human MSCs (hMSCs) were committed to differentiate when either adipogenic or osteogenic media was added, supplemented with five increasing concentrations of amitriptyline (0.001–10 µM), venlafaxine (0.01–25 µM), or fluoxetine (0.001–10 µM). Alizarin red staining (mineralization), alkaline phosphatase (osteoblastogenesis), and oil red O (adipogenesis) assays were performed at timed intervals. In addition, cell viability was assessed using a MTT. Results We found that fluoxetine had a significant inhibitory effect on mineralization. Furthermore, adipogenic differentiation of hMSC was affected by the addition of amitriptyline, venlafaxine, and fluoxetine to the media. Finally, none of the tested medications significantly affected cell survival. Conclusions This study showed a divergent effect of three antidepressants on hMSC differentiation, which appears to be independent of class and dose. As fluoxetine and amitriptyline, but not venlafaxine, affected both osteoblastogenesis and adipogenesis, this inhibitory effect could be associated to the high affinity of fluoxetine to the serotonin transporter system.
Collapse
Affiliation(s)
- Jeffrey S Kruk
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia
| | - Sandra Bermeo
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia.,Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Kristen K Skarratt
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia
| | - Stephen J Fuller
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia
| | - Gustavo Duque
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, Melbourne, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
49
|
Madrigal A, Tan L, Zhao Y. Expression regulation and functional analysis of RGS2 and RGS4 in adipogenic and osteogenic differentiation of human mesenchymal stem cells. Biol Res 2017; 50:43. [PMID: 29279050 PMCID: PMC5742872 DOI: 10.1186/s40659-017-0148-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 12/14/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Understanding the molecular basis underlying the formation of bone-forming osteocytes and lipid-storing adipocytes will help provide insights into the cause of disorders originating in stem/progenitor cells and develop therapeutic treatments for bone- or adipose-related diseases. In this study, the role of RGS2 and RGS4, two members of the regulators of G protein signaling (RGS) family, was investigated during adipogenenic and osteogenenic differentiation of human mesenchymal stem cells (hMSCs). RESULTS Expression of RGS2 and RGS4 were found to be inversely regulated during adipogenesis induced by dexamethasone (DEX) and 3-isobutyl-methylxanthine, regardless if insulin was present, with RGS2 up-regulated and RGS4 down-regulated in response to adipogenic induction. RGS2 expression was also up-regulated during osteogenesis at a level similar to that induced by treatment of DEX alone, a shared component of adipogenic and osteogenic differentiation inducing media, but significantly lower than the level induced by adipogenic inducing media. RGS4 expression was down-regulated during the first 48 h of osteogenesis but up-regulated afterwards, in both cases at levels similar to that induced by DEX alone. Expression knock-down using small interfering RNA against RGS2 resulted in decreased differentiation efficiency during both adipogenesis and osteogenesis. On the other hand, expression knock-down of RGS4 also resulted in decreased adipogenic differentiation but increased osteogenic differentiation. CONCLUSIONS RGS2 and RGS4 are differentially regulated during adipogenic and osteogenic differentiation of hMSCs. In addition, both RGS2 and RGS4 play positive roles during adipogenesis but opposing roles during osteogenesis, with RGS2 as a positive regulator and RGS4 as a negative regulator. These results imply that members of RGS proteins may play multifaceted roles during human adipogenesis and osteogenesis to balance or counterbalance each other's function during those processes.
Collapse
Affiliation(s)
- Alma Madrigal
- Biological Sciences Department, California State Polytechnic University at Pomona, 3801 W. Temple Ave., Pomona, CA, 91768, USA.,Center for Biomedicine and Genetics, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Lun Tan
- Biological Sciences Department, California State Polytechnic University at Pomona, 3801 W. Temple Ave., Pomona, CA, 91768, USA
| | - Yuanxiang Zhao
- Biological Sciences Department, California State Polytechnic University at Pomona, 3801 W. Temple Ave., Pomona, CA, 91768, USA.
| |
Collapse
|
50
|
Li JZ, Qu H, Wu J, Zhang F, Jia ZB, Sun JY, Lv B, Kang Y, Jiang SL, Kang K. Metabolic profiles of adipose-derived and bone marrow-derived stromal cells from elderly coronary heart disease patients by capillary liquid chromatography quadrupole time-of-flight mass spectrometry. Int J Mol Med 2017; 41:184-194. [PMID: 29115374 PMCID: PMC5746296 DOI: 10.3892/ijmm.2017.3198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 10/06/2017] [Indexed: 12/14/2022] Open
Abstract
Adipose-tissue derived mesenchymal stem cell (ADSC)-based therapy is a promising option for patients with atherosclerotic conditions, including coronary artery disease. However, the potential differences in the metabolic characteristics between bone marrow-derived mesenchymal stem cells (BMSCs) and ADSCs have remained to be fully elucidated. The present study aimed to compare the metabolic profiles of BMSCs and ADSCs via liquid chromatography quadrupole time-of-flight mass spectrometry. BMSCs and ADSCs obtained from elderly coronary heart disease patients were cultured, and after three passages, supernatants of each cell type were collected and systematically analysed. Substantial differences were detected between the metabolite signatures of ADSCs and BMSCs. In addition, further analysis using partial least-squares discriminant analysis score plots indicated significant differences between the supernatants of the two cell types. The following metabolites were deemed to be responsible for the potential differences in the metabolic characteristics of BMSCs and ADSCs: D-lactic acid, hydroxyindoleacetaldehyde, α-D-glucose, bovinic acid, 9,10-epoxyoctadecenoic acid, glyceraldehyde, phenylpyruvic acid, L-octanoylcarnitine, retinyl ester, α-ketoisovaleric acid, guanidoacetic acid, N-acetylneuraminic acid, imidazoleacetic acid riboside, sphingosine and pseudouridine 5′-phosphate. Based on these findings, there may be significant differences in the following metabolic pathways: The linoleic acid metabolic pathway, galactose metabolism, argentines and proline metabolism, retinol metabolism, glycine and serine metabolism, galactose metabolism, and amino sugar and nucleotide sugar metabolism. In conclusion, substantial differences in metabolic characteristics were detected between BMSCs and ADSCs, which may be associated with the different efficacies of atherosclerosis therapies employing these cell types.
Collapse
Affiliation(s)
- Jian-Zhong Li
- Key Laboratory of Education of the Ministry for Myocardial Ischemia, Division of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Hui Qu
- Key Laboratory of Education of the Ministry for Myocardial Ischemia, Division of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Jian Wu
- Key Laboratory of Education of the Ministry for Myocardial Ischemia, Division of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Fan Zhang
- Division of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Zhi-Bo Jia
- Key Laboratory of Education of the Ministry for Myocardial Ischemia, Division of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Jian-Yong Sun
- Division of Thoracic Surgery, Tang Du Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Bo Lv
- Key Laboratory of Education of the Ministry for Myocardial Ischemia, Division of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yue Kang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Shu-Lin Jiang
- Key Laboratory of Education of the Ministry for Myocardial Ischemia, Division of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Kai Kang
- Key Laboratory of Education of the Ministry for Myocardial Ischemia, Division of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|