1
|
Jaber M, Schmidt J, Kalkhof S, Gerstenfeld L, Duda GN, Checa S. OMIBONE: Omics-driven computer model of bone regeneration for personalized treatment. Bone 2024; 190:117288. [PMID: 39426580 DOI: 10.1016/j.bone.2024.117288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
Treatment of bone fractures are standardized according to the AO classification, which mainly refers to the mechanical stabilization required in a given situation but neglect individual differences due to patient's healing potential or accompanying diseases. Specially in elderly or immune-compromised patients, the complexity of individual constrains on a biological as well as mechanical level are hard to account for. Here, we introduce a novel framework that allows to predict bone regeneration outcome using combined proteomic and mechanical analyses in a computer model. The framework uses Ingenuity Pathway Analysis (IPA) software to link protein changes to alterations in biological processes and integrates these in an Agent-Based Model (ABM) of bone regeneration. This combined framework allows to predict bone formation and the potential of an individual to heal a given fracture setting. The performance of the framework was evaluated by replicating the experimental setup of a mouse femur fracture stabilized with an intramedullary pin. The model was informed by serum derived proteomics data. The tissue formation patterns were compared against experimental data based on x-ray and histology images. The results indicate the framework potential in predicting an individual's bone formation potential and hold promise as a concept to enable personalized bone healing predictions for a chosen fracture fixation.
Collapse
Affiliation(s)
- Mahdi Jaber
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - Johannes Schmidt
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Stefan Kalkhof
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Louis Gerstenfeld
- Department of Orthopaedic Surgery, Boston University of Medicine, Boston, MA, United States of America
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany.
| |
Collapse
|
2
|
Purbantoro SD, Taephatthanasagon T, Purwaningrum M, Hirankanokchot T, Peralta S, Fiani N, Sawangmake C, Rattanapuchpong S. Trends of regenerative tissue engineering for oral and maxillofacial reconstruction in veterinary medicine. Front Vet Sci 2024; 11:1325559. [PMID: 38450027 PMCID: PMC10915013 DOI: 10.3389/fvets.2024.1325559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Oral and maxillofacial (OMF) defects are not limited to humans and are often encountered in other species. Reconstructing significant tissue defects requires an excellent strategy for efficient and cost-effective treatment. In this regard, tissue engineering comprising stem cells, scaffolds, and signaling molecules is emerging as an innovative approach to treating OMF defects in veterinary patients. This review presents a comprehensive overview of OMF defects and tissue engineering principles to establish proper treatment and achieve both hard and soft tissue regeneration in veterinary practice. Moreover, bench-to-bedside future opportunities and challenges of tissue engineering usage are also addressed in this literature review.
Collapse
Affiliation(s)
- Steven Dwi Purbantoro
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Teeanutree Taephatthanasagon
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Medania Purwaningrum
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Thanyathorn Hirankanokchot
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Santiago Peralta
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Nadine Fiani
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Chenphop Sawangmake
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Sirirat Rattanapuchpong
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Academic Affairs, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
3
|
Han J, Li X, Liang B, Ma S, Pu Y, Yu F, Lu J, Ma Y, MacHugh DE, Jiang L. Transcriptome profiling of differentiating adipose-derived stem cells across species reveals new genes regulating adipogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159378. [PMID: 37572997 DOI: 10.1016/j.bbalip.2023.159378] [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: 04/11/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Adipose-derived stem cells (ADSCs) that are enriched in adipose tissue with multilineage differentiation potential have become an important tool in therapeutic research and tissue engineering. Certain breeds of sheep exhibit a unique fat tail trait such that tail tissue accounts for approximately 10 % of body weight and can provide an excellent source of ADSCs. Here, we describe isolation of primary ADSCs from ovine embryonic fat tail tissues that displayed high self-renewal capacity, multilineage differentiation and excellent adipogenic ability. Through transcriptome analysis covering ADSCs differentiating into adipocytes, 37 transcription factors were involved in early transcriptional events that initiate a regulatory cascade of adipogenesis; the entire adipogenic activity consists of a reduction in proliferation ability and upregulation of genes related to lipid generation and energy metabolism, as well as several genes associated with myogenesis. Furthermore, Comparative transcriptome analysis across species (sheep, human, and mouse) revealed enhanced basal metabolic ability in differentiating ovine ADSCs, which may relate to the excellent adipogenic capability of these cells. We also identified a small evolutionarily conserved gene set, consisting of 21 and 22 genes exhibiting increased and decreased expression, respectively. Almost half (20) of these genes have not previously been reported to regulate adipogenesis in mammals. In this study, we identified important regulators that trigger ovine adipocyte differentiation, main biological pathways involved in adipogenesis as well as the evolutionarily conserved genes governing adipogenic process across species. Our study provides a novel excellent biomaterial and novel genes regulating adipogenesis for cellular transplantation therapy and investigations of fat metabolism.
Collapse
Affiliation(s)
- Jiangang Han
- Laboratory of Animal Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China; Animal Genomics Laboratory, UCD School of Agriculture and Food Science, UCD College of Health and Agricultural Sciences, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
| | - Xiaojie Li
- Laboratory of Animal Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China; National Germplasm Center of Domestic Animal Resources, Ministry of Technology, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Benmeng Liang
- Laboratory of Animal Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China; National Germplasm Center of Domestic Animal Resources, Ministry of Technology, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Sijia Ma
- Laboratory of Animal Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China; Agricultural College, Ningxia University, Yinchuan, Ningxia, China
| | - Yabin Pu
- Laboratory of Animal Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China; National Germplasm Center of Domestic Animal Resources, Ministry of Technology, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Fuqing Yu
- National Animal Husbandry Service, Beijing 100193, China
| | - Jian Lu
- National Animal Husbandry Service, Beijing 100193, China
| | - Yuehui Ma
- Laboratory of Animal Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China; National Germplasm Center of Domestic Animal Resources, Ministry of Technology, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - David E MacHugh
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, UCD College of Health and Agricultural Sciences, University College Dublin, Belfield, Dublin D04 V1W8, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin D04 V1W8, Ireland.
| | - Lin Jiang
- Laboratory of Animal Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China; National Germplasm Center of Domestic Animal Resources, Ministry of Technology, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China.
| |
Collapse
|
4
|
Li S, Siengdee P, Oster M, Reyer H, Wimmers K, Ponsuksili S. Transcriptome changes during osteogenesis of porcine mesenchymal stem cells derived from different types of synovial membranes and genetic background. Sci Rep 2023; 13:10048. [PMID: 37344635 DOI: 10.1038/s41598-023-37260-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/19/2023] [Indexed: 06/23/2023] Open
Abstract
Synovial membrane mesenchymal stem cells (SMSCs) often serve as in vitro model for bone disease, but the molecular mechanisms driving osteogenesis in SMSCs from different donor cells of various sources and breeds remain unclear. In this study, porcine SMSCs isolated from adipose synovium (FP) and fibrous synovium (FS) of Angeln Saddleback (AS) and German Landrace (DL) were used to discover the signaling network change after osteogenic induction. During osteogenic differentiation, mineral deposition was first observed at day 14 and further increased until day 21. Transcriptional changes between day 1 and day 21 were enriched in several signaling pathways, including Wnt, PI3K-Akt, and TGF-beta pathway. Certain pathways related to osteogenesis, including osteoblast differentiation, regulation of bone mineralization, and BMP signaling pathway, were enriched at late time points, as confirmed by the osteogenic markers ALPL, COL1A1, and NANOG. A fraction of differentially expressed genes (DEGs) were found between FP and FS, while DEGs between AS and DL increased during the differentiation phase until day 7 and then decreased from day 14 to day 21. These genes are involved in several important signaling pathways, including TGF-beta, Wnt, and lipid-related signaling pathways, suggesting that SMSCs from these two breeds have different osteogenic capabilities.
Collapse
Affiliation(s)
- Shuaichen Li
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Puntita Siengdee
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
- Chulabhorn Graduate Institute, Program in Applied Biological Sciences, Chulabhorn Royal Academy, Kamphaeng Phet 6 Road, Laksi, Bangkok, 10210, Thailand
| | - Michael Oster
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Henry Reyer
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Klaus Wimmers
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
- Faculty of Agricultural and Environmental Sciences, University of Rostock, Justus-von-Liebig-Weg 6b, 18059, Rostock, Germany
| | - Siriluck Ponsuksili
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
5
|
Guo X, Xi L, Yu M, Fan Z, Wang W, Ju A, Liang Z, Zhou G, Ren W. Regeneration of articular cartilage defects: Therapeutic strategies and perspectives. J Tissue Eng 2023; 14:20417314231164765. [PMID: 37025158 PMCID: PMC10071204 DOI: 10.1177/20417314231164765] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/03/2023] [Indexed: 04/03/2023] Open
Abstract
Articular cartilage (AC), a bone-to-bone protective device made of up to 80% water and populated by only one cell type (i.e. chondrocyte), has limited capacity for regeneration and self-repair after being damaged because of its low cell density, alymphatic and avascular nature. Resulting repair of cartilage defects, such as osteoarthritis (OA), is highly challenging in clinical treatment. Fortunately, the development of tissue engineering provides a promising method for growing cells in cartilage regeneration and repair by using hydrogels or the porous scaffolds. In this paper, we review the therapeutic strategies for AC defects, including current treatment methods, engineering/regenerative strategies, recent advances in biomaterials, and present emphasize on the perspectives of gene regulation and therapy of noncoding RNAs (ncRNAs), such as circular RNA (circRNA) and microRNA (miRNA).
Collapse
Affiliation(s)
- Xueqiang Guo
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Lingling Xi
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Mengyuan Yu
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Zhenlin Fan
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Weiyun Wang
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Andong Ju
- Abdominal Surgical Oncology, Xinxiang
Central Hospital, Institute of the Fourth Affiliated Hospital of Xinxiang Medical
University, Xinxiang, China
| | - Zhuo Liang
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Guangdong Zhou
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
- Department of Plastic and
Reconstructive Surgery, Shanghai Key Lab of Tissue Engineering, Shanghai 9th
People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
- Guangdong Zhou, Department of Plastic and
Reconstructive Surgery, Shanghai Key Lab of Tissue Engineering, Shanghai 9th
People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639
Shanghai Manufacturing Bureau Road, Shanghai 200011, China.
| | - Wenjie Ren
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
- Wenjie Ren, Institute of Regenerative
Medicine and Orthopedics, Institutes of Health Central Plain, Xinxiang Medical
University, 601 Jinsui Avenue, Hongqi District, Xinxiang 453003, Henan, China.
| |
Collapse
|
6
|
Abstract
Bone is a living organ that exhibits active metabolic processes, presenting constant bone formation and resorption. The bone cells that maintain local homeostasis are osteoblasts, osteoclasts, osteocytes and bone marrow stem cells, their progenitor cells. Osteoblasts are the main cells that govern bone formation, osteoclasts are involved in bone resorption, and osteocytes, the most abundant bone cells, also participate in bone remodeling. All these cells have active metabolic activities, are interconnected and influence each other, having both autocrine and paracrine effects. Ageing is associated with multiple and complex bone metabolic changes, some of which are currently incompletely elucidated. Ageing causes important functional changes in bone metabolism, influencing all resident cells, including the mineralization process of the extracellular matrix. With advancing age, a decrease in bone mass, the appearance of specific changes in the local microarchitecture, a reduction in mineralized components and in load-bearing capacity, as well as the appearance of an abnormal response to different humoral molecules have been observed. The present review points out the most important data regarding the formation, activation, functioning, and interconnection of these bone cells, as well as data on the metabolic changes that occur due to ageing.
Collapse
Affiliation(s)
- Anca Cardoneanu
- Department of Rheumatology, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
- Clinical Rehabilitation Hospital, 1st Rheumatology Clinic, Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
- IIIrd Medical Clinic, "Saint Spiridon" Clinic Emergency County Hospital, Iasi, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Elena Rezus
- Department of Rheumatology, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
- Clinical Rehabilitation Hospital, 1st Rheumatology Clinic, Iasi, Romania
| |
Collapse
|
7
|
Lau CS, Chua J, Pena EM, Lim J, Saigo L, Goh BT. A Porcine Model Using Adipose Stem Cell-Loaded Scaffolds for Alveolar Ridge Augmentation. Tissue Eng Part C Methods 2022; 28:228-237. [PMID: 35442100 DOI: 10.1089/ten.tec.2022.0062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tooth loss greatly affects a person's quality of life and many turn to dental implants to replace lost teeth. The success of a dental implant depends on the amount of alveolar bone supporting the implant, and thus, bone augmentation is often necessary to preserve or build up bone volume in the alveolar ridge. Bone can be augmented with autogenous bone, allografts, or xenografts, but the limitations of such natural bone grafts prompt researchers to develop synthetic scaffolds supplemented with cells and/or bioactive agents as alternative bone grafts. The translation of these combination scaffolds from the laboratory to the clinic requires reliable experimental models that can simulate the clinical conditions in human patients. In this article, we describe the use of a porcine alveolar defect model as a platform to evaluate the efficacy of a novel combination of a three-dimensional-printed polycaprolactone-tricalcium phosphate (PCL-TCP) scaffold and adipose-derived mesenchymal stem cells (AD-MSCs) in lateral alveolar augmentation. The surgical protocol for the defect creation and regenerative surgery, as well as analytical methods to determine the extent of tissue regeneration, are described and discussed.
Collapse
Affiliation(s)
- Chau Sang Lau
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,National Dental Research Institute Singapore, National Dental Centre Singapore, Singapore, Singapore
| | - Jasper Chua
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Cardiovascular and Metabolic Disorder Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Edgar Macabe Pena
- SingHealth Experimental Medicine Centre and National Large Animal Research Facility, Singapore Health Services Pte Ltd., Singapore, Singapore
| | - Jing Lim
- Osteopore International Pte Ltd., Singapore, Singapore
| | - Leonardo Saigo
- Department of Oral and Maxillofacial Surgery, National Dental Centre Singapore, Singapore, Singapore
| | - Bee Tin Goh
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,National Dental Research Institute Singapore, National Dental Centre Singapore, Singapore, Singapore.,Department of Oral and Maxillofacial Surgery, National Dental Centre Singapore, Singapore, Singapore
| |
Collapse
|
8
|
Wu T, Tang H, Yang J, Yao Z, Bai L, Xie Y, Li Q, Xiao J. METTL3-m 6 A methylase regulates the osteogenic potential of bone marrow mesenchymal stem cells in osteoporotic rats via the Wnt signalling pathway. Cell Prolif 2022; 55:e13234. [PMID: 35470497 PMCID: PMC9136513 DOI: 10.1111/cpr.13234] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES Bone marrow mesenchymal stem cells (BMSCs) hold a high osteogenic differentiation potential, but the mechanisms that control the osteogenic ability of BMSCs from osteoporosis (OP-BMSCs) need further research. The purpose of this experiment is to discuss the osteogenic effect of Mettl3 on OP-BMSCs and explore new therapeutic target that can enhance the bone formation ability of OP-BMSCs. MATERIALS AND METHODS The bilateral ovariectomy (OVX) method was used to establish the SD rat OP model. Dot blots were used to reveal the different methylation levels of BMSCs and OP-BMSCs. Lentiviral-mediated overexpression of Mettl3 was applied in OP-BMSCs. QPCR and WB detected the molecular changes of osteogenic-related factors and Wnt signalling pathway in vitro experiment. The staining of calcium nodules and alkaline phosphatase detected the osteogenic ability of OP-BMSCs. Micro-CT and histological examination evaluated the osteogenesis of Mettl3 in OP rats in vivo. RESULTS The OP rat model was successfully established by OVX. Methylation levels and osteogenic potential of OP-BMSCs were decreased in OP-BMSCs. In vitro experiment, overexpression of Mettl3 could upregulate the osteogenic-related factors and activate the Wnt signalling pathway in OP-BMSCs. However, osteogenesis of OP-BMSCs was weakened by treatment with the canonical Wnt inhibitor Dickkopf-1. Micro-CT showed that the Mettl3(+) group had an increased amount of new bone formation at 8 weeks. Moreover, the results of histological staining were the same as the micro-CT results. CONCLUSIONS Taken together, the methylation levels and osteogenic potential of OP-BMSCs were decreased in OP-BMSCs. In vitro and in vivo studies, overexpression of Mettl3 could partially rescue the decreased bone formation ability of OP-BMSCs by the canonical Wnt signalling pathway. Therefore, Mettl3 may be a key targeted gene for bone generation and therapy of bone defects in OP patients.
Collapse
Affiliation(s)
- Tianli Wu
- Department of Oral ImplantologyThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
- Department of Oral and Maxillofacial SurgeryThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Hui Tang
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Jianghua Yang
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
- Department of Medical Technology, Faculty of Associated Medical SciencesChiang Mai UniversityChiang MaiThailand
| | - Zhihao Yao
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Long Bai
- Department of Oral ImplantologyThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Yuping Xie
- Department of Oral ImplantologyThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Qing Li
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Jingang Xiao
- Department of Oral ImplantologyThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
- Department of Oral and Maxillofacial SurgeryThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
9
|
Liu Y, Wang Z, Ju M, Zhao Y, Jing Y, Li J, Shao C, Fu T, Lv Z, Li G. Modification of COL1A1 in Autologous Adipose Tissue-Derived Progenitor Cells Rescues the Bone Phenotype in a Mouse Model of Osteogenesis Imperfecta. J Bone Miner Res 2021; 36:1521-1534. [PMID: 33950576 DOI: 10.1002/jbmr.4326] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/17/2022]
Abstract
Osteogenesis imperfecta (OI) is a congenital genetic disorder mainly manifested as bone fragility and recurrent fracture. Mutation of COL1A1/COL1A2 genes encoding the type I collagen are most responsible for the clinical patients. Allogenic mesenchymal stem cells (MSCs) provide the potential to treat OI through differentiation into osteoblasts. Autologous defective MSCs have not been utilized in OI treatment mainly because of their impaired osteogenesis, but the latent mechanism has not been well understood. Here, the relative signaling abnormality of adipose-derived mesenchymal stem cells (ADSCs) isolated from OI type I mice (Col1a1+/-365 mice) was explored. Autologous ADSCs transfected by retrovirus carrying human COL1A1 gene was first utilized in OI therapy. The results showed that decreased activity of Yes-associated protein (YAP) due to hyperactive upstream Hippo kinases greatly contributed to the weakened bone-forming capacity of defective ADSCs. Recovered collagen synthesis of autologous ADSCs by COL1A1 gene modification normalized Hippo/YAP signaling and effectively rescued YAP-mediated osteogenesis. And the COL1A1 gene engineered autologous ADSCs efficaciously improved the microstructure, enhanced the mechanical properties and promoted bone formation of Col1a1+/-365 mice after femoral bone marrow cavity delivery and might serve as an alternative source of stem cells in OI treatment. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Yi Liu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Zihan Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Mingyan Ju
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yuxia Zhao
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yaqing Jing
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Jiaci Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Chenyi Shao
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Ting Fu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Zhe Lv
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Guang Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
10
|
Genome-wide identification and characterization of perirenal adipose tissue microRNAs in rabbits fed a high-fat diet. Biosci Rep 2021; 41:228333. [PMID: 33851695 PMCID: PMC8082595 DOI: 10.1042/bsr20204297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 01/03/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of endogenous single-stranded RNA molecules that play an important role in gene regulation in animals by pairing with target gene mRNA. Extensive evidence shows that miRNAs are key players in metabolic regulation and the development of obesity. However, the systemic understanding of miRNAs in the adipogenesis of obese rabbits need further investigation. Here, seven small RNA libraries from rabbits fed either a standard normal diet (SND; n=3) or high-fat diet (HFD; n=4) were constructed and sequenced. Differentially expressed (DE) miRNAs were identified using the edgeR data analysis package from R. Software miRanda and RNAhybrid were used to predict the target genes of miRNAs. To further explore the functions of DE miRNAs, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed. A total of 81449996 clean reads were obtained from the seven libraries, of which, 52 known DE miRNAs (24 up-regulated, 28 down-regulated) and 31 novel DE miRNAs (14 up-regulated, 17 down-regulated) were identified. GO enrichment analysis revealed that the DE miRNAs target genes were involved in intermediate filament cytoskeleton organization, intermediate filament-based process, and α-tubulin binding. DE miRNAs were involved in p53 signaling, linoleic acid metabolism, and other adipogenesis-related KEGG pathways. Our study further elucidates the possible functions of DE miRNAs in rabbit adipogenesis, contributing to the understanding of rabbit obesity.
Collapse
|
11
|
Shao J, Bai X, Pan T, Li Y, Jia X, Wang J, Lai S. Genome-Wide DNA Methylation Changes of Perirenal Adipose Tissue in Rabbits Fed a High-Fat Diet. Animals (Basel) 2020; 10:E2213. [PMID: 33255930 PMCID: PMC7761299 DOI: 10.3390/ani10122213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/22/2022] Open
Abstract
DNA methylation is an epigenetic mechanism that plays an important role in gene regulation without an altered DNA sequence. Previous studies have demonstrated that diet affects obesity by partially mediating DNA methylation. Our study investigated the genome-wide DNA methylation of perirenal adipose tissue in rabbits to identify the epigenetic changes of high-fat diet-mediated obesity. Two libraries were constructed pooling DNA of rabbits fed a standard normal diet (SND) and DNA of rabbits fed a high-fat diet (HFD). Differentially methylated regions (DMRs) were identified using the option of the sliding window method, and online software DAVID Bioinformatics Resources 6.7 was used to perform Gene Ontology (GO) terms and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analysis of DMRs-associated genes. A total of 12,230 DMRs were obtained, of which 2305 (1207 up-regulated, 1098 down-regulated) and 601 (368 up-regulated, 233 down-regulated) of identified DMRs were observed in the gene body and promoter regions, respectively. GO analysis revealed that the DMRs-associated genes were involved in developmental process (GO:0032502), cell differentiation (GO:0030154), and lipid binding (GO:0008289), and KEGG pathway enrichment analysis revealed the DMRs-associated genes were enriched in linoleic acid metabolism (KO00591), DNA replication (KO03030), and MAPK signaling pathway (KO04010). Our study further elucidates the possible functions of DMRs-associated genes in rabbit adipogenesis, contributing to the understanding of HFD-mediated obesity.
Collapse
Affiliation(s)
- Jiahao Shao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (X.B.); (Y.L.); (X.J.); (J.W.)
| | - Xue Bai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (X.B.); (Y.L.); (X.J.); (J.W.)
| | - Ting Pan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China;
| | - Yanhong Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (X.B.); (Y.L.); (X.J.); (J.W.)
| | - Xianbo Jia
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (X.B.); (Y.L.); (X.J.); (J.W.)
| | - Jie Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (X.B.); (Y.L.); (X.J.); (J.W.)
| | - Songjia Lai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (X.B.); (Y.L.); (X.J.); (J.W.)
| |
Collapse
|
12
|
Han S, Kim J, Lee G, Kim D. Mechanical Properties of Materials for Stem Cell Differentiation. ACTA ACUST UNITED AC 2020; 4:e2000247. [DOI: 10.1002/adbi.202000247] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/28/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Seong‐Beom Han
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| | - Jeong‐Ki Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| | - Geonhui Lee
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| | - Dong‐Hwee Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| |
Collapse
|
13
|
Filho DM, de Carvalho Ribeiro P, Oliveira LF, Dos Santos ALRT, Parreira RC, Pinto MCX, Resende RR. Enhancing the Therapeutic Potential of Mesenchymal Stem Cells with the CRISPR-Cas System. Stem Cell Rev Rep 2020; 15:463-473. [PMID: 31147819 DOI: 10.1007/s12015-019-09897-0] [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] [Indexed: 12/24/2022]
Abstract
Mesenchymal stem cells (MSCs), also known as multipotent mesenchymal stromal stem cells, are found in the perivascular space of several tissues. These cells have been subject of intense research in the last decade due to their low teratogenicity, as well as their ability to differentiate into mature cells and to secrete immunomodulatory and trophic factors. However, they usually promote only a modest benefit when transplanted in experimental disease models, one of the limitations for their clinical application. The CRISPR-Cas system, in turn, is highlighted as a simple and effective tool for genetic engineering. This system was tested in clinical trials over a relatively short period of time after establishing its applicability to the edition of the mammalian cell genome. Similar to the research evolution in MSCs, the CRISPR-Cas system demonstrated inconsistencies that limited its clinical application. In this review, we outline the evolution of MSC research and its applicability, and the progress of the CRISPR-Cas system from its discovery to the most recent clinical trials. We also propose perspectives on how the CRISPR-Cas system may improve the therapeutic potential of MSCs, making it more beneficial and long lasting.
Collapse
Affiliation(s)
- Daniel Mendes Filho
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Patrícia de Carvalho Ribeiro
- Laboratory of Immunology and Experimental Transplantation, São José do Rio Preto Medical School, São José do Rio Preto, São Paulo, Brazil.,Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Lucas Felipe Oliveira
- Department of Physiology, Biological and Natural Sciences Institute, Triangulo Mineiro Federal University, Uberaba, Minas Gerais, Brazil.,National Institute of Science and Technology for Regenerative Medicine (INCT-REGENERA-CNPq), Rio de Janeiro, RJ, Brazil.,Minas Gerais Network for Tissue Engineering and Cell Therapy (REMETTECFAPEMIG), Belo Horizonte, MG, Brazil
| | | | - Ricardo Cambraia Parreira
- Department of Pharmacology, Biological Sciences Institute, Goias Federal University, Goiania, Goias, Brazil.
| | - Mauro Cunha Xavier Pinto
- Department of Pharmacology, Biological Sciences Institute, Goias Federal University, Goiania, Goias, Brazil
| | - Rodrigo Ribeiro Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
14
|
Corrado A, Cici D, Rotondo C, Maruotti N, Cantatore FP. Molecular Basis of Bone Aging. Int J Mol Sci 2020; 21:ijms21103679. [PMID: 32456199 PMCID: PMC7279376 DOI: 10.3390/ijms21103679] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/16/2022] Open
Abstract
A decline in bone mass leading to an increased fracture risk is a common feature of age-related bone changes. The mechanisms underlying bone senescence are very complex and implicate systemic and local factors and are the result of the combination of several changes occurring at the cellular, tissue and structural levels; they include alterations of bone cell differentiation and activity, oxidative stress, genetic damage and the altered responses of bone cells to various biological signals and to mechanical loading. The molecular mechanisms responsible for these changes remain greatly unclear and many data derived from in vitro or animal studies appear to be conflicting and heterogeneous, probably due to the different experimental approaches; nevertheless, understanding the main physio-pathological processes that cause bone senescence is essential for the development of new potential therapeutic options for treating age-related bone loss. This article reviews the current knowledge concerning the molecular mechanisms underlying the pathogenesis of age-related bone changes.
Collapse
|
15
|
Senile Osteoporosis: The Involvement of Differentiation and Senescence of Bone Marrow Stromal Cells. Int J Mol Sci 2020; 21:ijms21010349. [PMID: 31948061 PMCID: PMC6981793 DOI: 10.3390/ijms21010349] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 12/26/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022] Open
Abstract
Senile osteoporosis has become a worldwide bone disease with the aging of the world population. It increases the risk of bone fracture and seriously affects human health. Unlike postmenopausal osteoporosis which is linked to menopause in women, senile osteoporosis is due to aging, hence, affecting both men and women. It is commonly found in people with more than their 70s. Evidence has shown that with age increase, bone marrow stromal cells (BMSCs) differentiate into more adipocytes rather than osteoblasts and undergo senescence, which leads to decreased bone formation and contributes to senile osteoporosis. Therefore, it is necessary to uncover the molecular mechanisms underlying the functional changes of BMSCs. It will benefit not only for understanding the senile osteoporosis development, but also for finding new therapies to treat senile osteoporosis. Here, we review the recent advances of the functional alterations of BMSCs and the related mechanisms during senile osteoporosis development. Moreover, the treatment of senile osteoporosis by aiming at BMSCs is introduced.
Collapse
|
16
|
Minuti A, Bionaz M, Lopreiato V, Janovick NA, Rodriguez-Zas SL, Drackley JK, Loor JJ. Prepartum dietary energy intake alters adipose tissue transcriptome profiles during the periparturient period in Holstein dairy cows. J Anim Sci Biotechnol 2020; 11:1. [PMID: 31908775 PMCID: PMC6941259 DOI: 10.1186/s40104-019-0409-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background The aim of the study was to investigate the effect of energy overfeeding during the dry period on adipose tissue transcriptome profiles during the periparturient period in dairy cows. Methods Fourteen primiparous Holstein cows from a larger cohort receiving a higher-energy diet (1.62 Mcal of net energy for lactation/kg of dry matter; 15% crude protein) for ad libitum intake to supply 150% (OVR) or 100% (CTR) of energy requirements from dry off until parturition were used. After calving, all cows received the same lactation diet. Subcutaneous adipose tissue (SAT) biopsies were collected at - 14, 1, and 14 d from parturition (d) and used for transcriptome profiling using a bovine oligonucleotide microarray. Data mining of differentially expressed genes (DEG) between treatments and due to sampling time was performed using the Dynamic Impact Approach (DIA) and Ingenuity Pathway Analysis (IPA). Results There was a strong effect of over-feeding energy on DEG with 2434 (False discovery rate-corrected P < 0.05) between OVR and CTR at - 14 d, and only 340 and 538 at 1 and 14 d. The most-impacted and activated pathways in the Kyoto Encyclopedia of Genes and Genomes (KEGG) database that were highlighted by DIA analysis at - 14 d in OVR vs. CTR included 9 associated with carbohydrate metabolism, with 'Pyruvate metabolism', 'Glycolysis/gluconeogenesis', and 'Pentose phosphate pathway' among the most-activated. Not surprisingly, OVR led to marked activation of lipid metabolism (e.g. 'Fatty acid biosynthesis' and 'Glycerolipid metabolism'). Unexpected metabolic pathways that were activated at - 14 d in OVR included several related to metabolism of amino acids (e.g. branched chain) and of cofactors and vitamins (thiamin). Among endocrine and immune system pathways, at - 14 d OVR led to marked activation of 'PPAR signalling' and 'Antigen processing and presentation'. Among key pathways affected over time in OVR, a number were related to translation (e.g. mTOR signaling), endocrine/immune signaling (CXCR4 and IGF1), and lipid metabolism (oxidative phosphorylation) with greater activation in OVR vs. CTR specifically at - 14 d. Although statistical differences for several pathways in OVR vs. CTR nearly disappeared at 1 and 14 vs. - 14 d, despite the well-known catabolic state of adipose depots after calving, the bioinformatics analyses suggested important roles for a number of signaling mechanisms at - 14 vs. 14 than 1 vs. -14 d. This was particularly evident in cows fed to meet predicted energy requirements during the dry period (CTR). Conclusions Data underscored a strong activation by overfeeding energy of anabolic processes in the SAT exclusively prepartum. The study confirmed that higher-energy diets prepartum drive a transcriptional cascade of events orchestrated in part by the activation of PPARγ that regulate preadipocyte differentiation and lipid storage in SAT. Novel aspects of SAT biology to energy overfeeding or change in physiologic state also were uncovered, including the role of amino acid metabolism, mTOR signaling, and the immune system.
Collapse
Affiliation(s)
- Andrea Minuti
- 1Department of Animal Sciences,Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Massimo Bionaz
- 2Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97330 USA
| | - Vincenzo Lopreiato
- 1Department of Animal Sciences,Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Nicole A Janovick
- 3Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Sandra L Rodriguez-Zas
- 3Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - James K Drackley
- 3Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Juan J Loor
- 3Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| |
Collapse
|
17
|
Marcon BH, Spangenberg L, Bonilauri B, Robert AW, Angulski ABB, Cabo GC, Cofré AR, Bettes PSL, Dallagiovanna B, Shigunov P. Data describing the experimental design and quality control of RNA-Seq of human adipose-derived stem cells undergoing early adipogenesis and osteogenesis. Data Brief 2019; 28:105053. [PMID: 31989002 PMCID: PMC6970145 DOI: 10.1016/j.dib.2019.105053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/22/2019] [Accepted: 12/17/2019] [Indexed: 11/28/2022] Open
Abstract
An important tool to study the regulation of gene expression is the sequencing and the analysis of different RNA fractions: total, ribosome-free, monosomal and polysomal. By comparing these different populations, it is possible to identity which genes are differentially expressed and to get information on how transcriptional and translational regulation modulates cellular function. Therefore, we used this strategy to analyze the regulation of gene expression of human adipose-derived stem cells during the triggering of the adipogenic and osteogenic differentiation. Here, we have focused on analyzing the differential expression of mRNAs during early adipogenic and osteogenic differentiation, and presented the detailed data concerning the experimental design, the RNA-Seq quality data, the raw data obtained and the RT-qPCR validation data. This information is important to confirm the accuracy of the data considering a future reuse of the data provided. Moreover, this study may be used as groundwork for future characterization of the transcriptome and the translatome regulation of different cell types.
Collapse
Affiliation(s)
- Bruna H Marcon
- Laboratory of Basic Biology of Stem Cells (LABCET), Instituto Carlos Chagas - FIOCRUZ-PR, Curitiba, Paraná, 81350-010, Brazil
| | - Lucia Spangenberg
- Unidad de Bioinformática, Institut Pasteur Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay
| | - Bernardo Bonilauri
- Laboratory of Basic Biology of Stem Cells (LABCET), Instituto Carlos Chagas - FIOCRUZ-PR, Curitiba, Paraná, 81350-010, Brazil
| | - Anny Waloski Robert
- Laboratory of Basic Biology of Stem Cells (LABCET), Instituto Carlos Chagas - FIOCRUZ-PR, Curitiba, Paraná, 81350-010, Brazil
| | - Addeli Bez Batti Angulski
- Laboratory of Basic Biology of Stem Cells (LABCET), Instituto Carlos Chagas - FIOCRUZ-PR, Curitiba, Paraná, 81350-010, Brazil
| | - Guillermo Cabrera Cabo
- Unidad de Bioinformática, Institut Pasteur Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay
| | - Axel R Cofré
- Laboratory of Basic Biology of Stem Cells (LABCET), Instituto Carlos Chagas - FIOCRUZ-PR, Curitiba, Paraná, 81350-010, Brazil
| | | | - Bruno Dallagiovanna
- Laboratory of Basic Biology of Stem Cells (LABCET), Instituto Carlos Chagas - FIOCRUZ-PR, Curitiba, Paraná, 81350-010, Brazil
| | - Patrícia Shigunov
- Laboratory of Basic Biology of Stem Cells (LABCET), Instituto Carlos Chagas - FIOCRUZ-PR, Curitiba, Paraná, 81350-010, Brazil
| |
Collapse
|
18
|
Vargas-Bello-Pérez E, Bionaz M, Sciarresi-Arechabala P, Cancino-Padilla N, Morales MS, Romero J, Leskinen H, Garnsworthy PC, Loor JJ. Long-Term Effects of Dietary Olive Oil and Hydrogenated Vegetable Oil on Expression of Lipogenic Genes in Subcutaneous Adipose Tissue of Dairy Cows. Vet Sci 2019; 6:vetsci6030074. [PMID: 31540163 PMCID: PMC6789855 DOI: 10.3390/vetsci6030074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 12/25/2022] Open
Abstract
The objective of this study was to characterize the long-term transcriptomic effects of lipogenic genes in subcutaneous adipose tissue (SAT) of dairy cows supplemented with unsaturated (olive oil; OO) and saturated (hydrogenated vegetable oil; HVO) lipids. Cows were fed a control diet with no added lipid, or diets containing OO or HVO (n = 5 cows/group) for 63 days. SAT was obtained from the tail-head area at the onset of the study and after 21, 42, and 63 days of supplementation. Treatments had minor effects on expression of measured genes. Both fat supplements reduced expression of PPARG, HVO decreased transcription of the desaturase FADS2 and lipid droplet formation PLIN2, and OO increased transcription of FABP3. Both lipid treatments decreased expression of the transcription regulator SREBF1 and its chaperone (SCAP) during the first 21 days of treatment. Our data indicated that long-term feeding of OO and HVO have a relatively mild effect on expression of lipogenic genes in SAT of mid-lactating cows.
Collapse
Affiliation(s)
- Einar Vargas-Bello-Pérez
- Departamento de Ciencias Animales, Facultad de Agronomía e Ingeniería Forestal, Pontificia Universidad Católica de Chile, Santiago Casilla-306, Chile.
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegardsvej 3, C DK-1870 Frederiksberg, Denmark.
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | - Pietro Sciarresi-Arechabala
- Departamento de Fomento de la Producción Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile. Av. Santa Rosa 11735, La Pintana, Santiago 8820808, Chile.
| | - Nathaly Cancino-Padilla
- Departamento de Ciencias Animales, Facultad de Agronomía e Ingeniería Forestal, Pontificia Universidad Católica de Chile, Santiago Casilla-306, Chile.
| | - María Sol Morales
- Departamento de Fomento de la Producción Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile. Av. Santa Rosa 11735, La Pintana, Santiago 8820808, Chile.
| | - Jaime Romero
- Instituto de Nutrición y Tecnología de los Alimentos, Santiago 7810000, Chile.
| | - Heidi Leskinen
- Milk Production, Production Systems, Natural Resources Institute Finland (Luke), 31600 Jokioinen FI, Finland.
| | - Philip C Garnsworthy
- School of Biosciences, The University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK.
| | - Juan J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, Mammalian NutriPhysioGenomics, University of Illinois, Urbana, IL 61801, USA.
| |
Collapse
|
19
|
Chen Y, Cao J, Zhang N, Yang B, He Q, Shao X, Ying M. Advances in differentiation therapy for osteosarcoma. Drug Discov Today 2019; 25:497-504. [PMID: 31499188 DOI: 10.1016/j.drudis.2019.08.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/04/2019] [Accepted: 08/28/2019] [Indexed: 02/08/2023]
Abstract
Differentiation therapy involves the use of agents that can induce differentiation in cancer cells, with the irreversible loss of tumour phenotype. The application of differentiation therapy in osteosarcoma has made progress because of a better understanding of the potential links between differentiation defects and tumorigenesis. Here, we review recent studies on differentiation therapy for osteosarcoma, describing a variety of differentiation inducers. By highlighting these examples of drug-induced osteosarcoma cell differentiation, we can acquire unique insights into not only osteosarcoma treatment, but also novel approaches to transform differentiating drugs into more effective therapies for other solid tumours.
Collapse
Affiliation(s)
- Yingqian Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ning Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xuejing Shao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
20
|
Stem cells in Osteoporosis: From Biology to New Therapeutic Approaches. Stem Cells Int 2019; 2019:1730978. [PMID: 31281368 PMCID: PMC6589256 DOI: 10.1155/2019/1730978] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis is a systemic disease that affects the skeleton, causing reduction of bone density and mass, resulting in destruction of bone microstructure and increased risk of bone fractures. Since osteoporosis is a disease affecting the elderly and the aging of the world's population is constantly increasing, it is expected that the incidence of osteoporosis and its financial burden on the insurance systems will increase continuously and there is a need for more understanding this condition in order to prevent and/or treat it. At present, available drug therapy for osteoporosis primarily targets the inhibition of bone resorption and agents that promote bone mineralization, designed to slow disease progression. Safe and predictable pharmaceutical means to increase bone formation have been elusive. Stem cell therapy of osteoporosis, as a therapeutic strategy, offers the promise of an increase in osteoblast differentiation and thus reversing the shift towards bone resorption in osteoporosis. This review is focused on the current views regarding the implication of the stem cells in the cellular and physiologic mechanisms of osteoporosis and discusses data obtained from stem cell-based therapies of osteoporosis in experimental animal models and the possibility of their future application in clinical trials.
Collapse
|
21
|
Dewey MJ, Johnson EM, Weisgerber DW, Wheeler MB, Harley BAC. Shape-fitting collagen-PLA composite promotes osteogenic differentiation of porcine adipose stem cells. J Mech Behav Biomed Mater 2019; 95:21-33. [PMID: 30953806 DOI: 10.1016/j.jmbbm.2019.03.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/17/2018] [Accepted: 03/17/2019] [Indexed: 10/27/2022]
Abstract
Craniomaxillofacial bone defects can occur as a result of congenital, post-oncologic, and high-energy impact conditions. The scale and irregularity of such defects motivate new biomaterials to promote regeneration of the damaged bone. We have recently described a mineralized collagen scaffold capable of instructing stem cell osteogenic differentiation and new bone infill in the absence of traditional osteogenic supplements. Herein, we report the integration of a millimeter-scale reinforcing poly (lactic acid) frame fabricated via 3D-printing into the mineralized collagen scaffold with micron-scale porosity to form a multi-scale mineralized collagen-PLA composite. We describe modifications to the PLA frame design to increase the compressive strength (Young's Modulus, ultimate stress and strain) of the composite. A critical challenge beyond increasing the compressive strength of the collagen scaffold is addressing challenges inherent with the irregularity of clinical defects. As a result, we examined the potential for modifying the frame architecture to render the composite with increased compressive strength in one axis or radial compressibility and shape-fitting capacity in an orthogonal axis. A library of mineralized collagen-PLA composites was mechanically characterized via compression testing and push-out test to describe mechanical performance and shape-fitting capacity. We also report in vitro comparison of the bioactivity of porcine adipose derived stem cells in the mineralized collagen-PLA composite versus the mineralized collagen scaffold via metabolic activity, gene expression, and functional matrix synthesis. The results suggest that incorporation of the PLA reinforcing frame does not negatively influence the osteoinductive nature of the mineralized collagen scaffold. Together, these findings suggest a strategy to address often competing bioactivity, mechanical strength, and shape-fitting design requirements for biomaterials for craniomaxillofacial bone regeneration.
Collapse
Affiliation(s)
- Marley J Dewey
- Dept. of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Eileen M Johnson
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Daniel W Weisgerber
- Dept. of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Matthew B Wheeler
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Brendan A C Harley
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Dept. of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
22
|
Smieszek A, Tomaszewski KA, Kornicka K, Marycz K. Metformin Promotes Osteogenic Differentiation of Adipose-Derived Stromal Cells and Exerts Pro-Osteogenic Effect Stimulating Bone Regeneration. J Clin Med 2018; 7:E482. [PMID: 30486321 PMCID: PMC6306720 DOI: 10.3390/jcm7120482] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/12/2018] [Accepted: 11/22/2018] [Indexed: 02/07/2023] Open
Abstract
Metformin, the gold standard in type 2 diabetes treatment, is a drug with multi-faceted effects. Currently, metformin has gained much attention as an agent that may find application in regenerative medicine. In this study, we considered its pro-osteogenic function in the course of in vitro osteogenesis of multipotent stromal cells derived from rat adipose tissue (rASCs). In addition, we evaluated the effect of metformin treatment on bone metabolism in a model of cranial defect in nondiabetic rats. In vitro study showed that metformin that is introduced to the culture medium at concentration equal 500 µM may promote the differentiation of rASCs into bone-forming cells, which express mRNA and secrets proteins that are related to the functional tissue (namely, alkaline phosphatase and osteocalcin). Osteogenic effect of metformin, as determined using in vitro model, was also manifested with the formation of mineralized extracellular matrix rich calcium and phosphorous deposits. We have also found, that in undifferentiated rASCs, metformin significantly activates a critical regulatory factor for osteogenic differentiation, i.e., AMPK. Moreover, using in vivo model we showed metformin administration at a dose of 250 mg/kg/day accelerated bone healing and the formation of mature tissue at a fracture site in rat cranial defect model. The obtained results shed promising light on metformin application in regenerative orthopedics, both as an agent improving functionality of ASCs for therapeutic transplantation, as well as a medication enhancing the bone healing process.
Collapse
Affiliation(s)
- Agnieszka Smieszek
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences Wroclaw 50-375, Poland.
| | - Krzysztof A Tomaszewski
- Department of Anatomy, Jagiellonian University Medical College, 12 Kopernika Street, 31-034 Krakow, Poland.
| | - Katarzyna Kornicka
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences Wroclaw 50-375, Poland.
| | - Krzysztof Marycz
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences Wroclaw 50-375, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392 Gießen, Germany.
| |
Collapse
|
23
|
Luo G, Xu B, Wang W, Wu Y, Li M. Study of the osteogenesis effect of icariside II and icaritin on canine bone marrow mesenchymal stem cells. J Bone Miner Metab 2018; 36:668-678. [PMID: 29264750 DOI: 10.1007/s00774-017-0889-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 11/15/2017] [Indexed: 10/18/2022]
Abstract
This study aimed to identify the osteogenesis effect of icariside II (ICSII) and icaritin (ICT) in vitro. Bone marrow mesenchymal stem cells (BMSCs) were treated with ICSII and ICT in order to detect the proliferation and differentiation of BMSCs, the expression of the osteogenesis-related proteins with or without osteogenic medium (OM) and genes, Runt-related transcription factor 2 (Runx-2), osteocalcin (OCN), osteopontin (OPN), osterix, and basic fibroblast growth factor (bFGF), and the phosphorylation levels of mitogen-activated protein kinase (MAPK). We found that the optical density increased and alkaline phosphatase decreased after the BMSCs were treated with different concentrations of ICSII; however, ICT showed an opposing effect. The formation of calcium nodules was observed after the BMSCs were treated with ICSII and ICT. The expression level of osteogenesis-related proteins was enhanced following treatment with both ICSII or ICT, while the expression level of the osteogenesis-related genes Runx-2, OCN, OPN, osterix, and bFGF significantly increased with ICSII treatment (P < 0.05), and only Runx-2 and bFGF significantly increased (P < 0.01) with ICT. The expression of osteogenic differentiation-related proteins (except OPN) following treatment with ICSII + OM or ICT + OM was not notably increased. Both ICSII and ICT elevated the phosphorylation levels of MAPK/ERK, which was attenuated by GDC-0994 (an inhibitor of MAPK/ERK). Collectively, these data indicate that ICSII and ICT facilitate orientation osteogenic differentiation of BMSCs, which is most likely via the MAPK/ERK pathway. OM did not synergistically enhance the osteogenesis effect of ICSII and ICT.
Collapse
Affiliation(s)
- Guangming Luo
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Block C No 1088 of Hai Yuan Road, High and New Technology Zone, Kunming, 650031, Yunnan, People's Republic of China.
| | - Biao Xu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Block C No 1088 of Hai Yuan Road, High and New Technology Zone, Kunming, 650031, Yunnan, People's Republic of China
| | - Weihong Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Block C No 1088 of Hai Yuan Road, High and New Technology Zone, Kunming, 650031, Yunnan, People's Republic of China
| | - Yong Wu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Block C No 1088 of Hai Yuan Road, High and New Technology Zone, Kunming, 650031, Yunnan, People's Republic of China
| | - Ming Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Block C No 1088 of Hai Yuan Road, High and New Technology Zone, Kunming, 650031, Yunnan, People's Republic of China
| |
Collapse
|
24
|
Qiu K, Zhang X, Wang L, Jiao N, Xu D, Yin J. Protein Expression Landscape Defines the Differentiation Potential Specificity of Adipogenic and Myogenic Precursors in the Skeletal Muscle. J Proteome Res 2018; 17:3853-3865. [DOI: 10.1021/acs.jproteome.8b00530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kai Qiu
- State Key Lab of Animal Nutrition & Ministry of Agriculture Feed Industry Centre, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China
| | - Xin Zhang
- State Key Lab of Animal Nutrition & Ministry of Agriculture Feed Industry Centre, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China
| | - Liqi Wang
- State Key Lab of Animal Nutrition & Ministry of Agriculture Feed Industry Centre, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China
| | - Ning Jiao
- State Key Lab of Animal Nutrition & Ministry of Agriculture Feed Industry Centre, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China
| | - Doudou Xu
- State Key Lab of Animal Nutrition & Ministry of Agriculture Feed Industry Centre, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China
| | - Jingdong Yin
- State Key Lab of Animal Nutrition & Ministry of Agriculture Feed Industry Centre, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
25
|
Sadie-Van Gijsen H. Adipocyte biology: It is time to upgrade to a new model. J Cell Physiol 2018; 234:2399-2425. [PMID: 30192004 DOI: 10.1002/jcp.27266] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/25/2018] [Indexed: 12/15/2022]
Abstract
Globally, the obesity pandemic is profoundly affecting quality of life and economic productivity, but efforts to address this, especially on a pharmacological level, have generally proven unsuccessful to date, serving as a stark demonstration that our understanding of adipocyte biology and pathophysiology is incomplete. To deliver better insight into adipocyte function and obesity, we need improved adipocyte models with a high degree of fidelity in representing the in vivo state and with a diverse range of experimental applications. Adipocyte cell lines, especially 3T3-L1 cells, have been used extensively over many years, but these are limited in terms of relevance and versatility. In this review, I propose that primary adipose-derived stromal/stem cells (ASCs) present a superior model with which to study adipocyte biology ex vivo. In particular, ASCs afford us the opportunity to study adipocytes from different, functionally distinct, adipose depots and to investigate, by means of in vivo/ex vivo studies, the effects of many different physiological and pathophysiological factors, such as age, body weight, hormonal status, diet and nutraceuticals, as well as disease and pharmacological treatments, on the biology of adipocytes and their precursors. This study will give an overview of the characteristics of ASCs and published studies utilizing ASCs, to highlight the areas where our knowledge is lacking. More comprehensive studies in primary ASCs will contribute to an improved understanding of adipose tissue, in healthy and dysfunctional states, which will enhance our efforts to more successfully manage and treat obesity.
Collapse
Affiliation(s)
- Hanél Sadie-Van Gijsen
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa.,Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa
| |
Collapse
|
26
|
Romeo L, Diomede F, Gugliandolo A, Scionti D, Lo Giudice F, Lanza Cariccio V, Iori R, Bramanti P, Trubiani O, Mazzon E. Moringin Induces Neural Differentiation in the Stem Cell of the Human Periodontal Ligament. Sci Rep 2018; 8:9153. [PMID: 29904155 PMCID: PMC6002387 DOI: 10.1038/s41598-018-27492-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/04/2018] [Indexed: 12/26/2022] Open
Abstract
The therapeutic strategies for neurodegenerative diseases still represent a vast research field because of the lack of targeted, effective and resolutive treatment for neurodegenerative diseases. The use of stem cell-based therapy is an alternative approach that could lead to the replacement of damaged neuronal tissue. For this purpose, adult mesenchymal stem cells (MSC), including periodontal ligament stem cells (PDLSCs), could be very useful for their differentiation capacity, easy isolation and the ability to perform an autologous implant. The aim of this work was to test whether the Moringin [4-(α-L-rhamnosyloxy) benzyl isothiocyanate; GMG-ITC], an isothiocyanate extracted from Moringa oleifera seeds, was able to induce PDLSCs toward neural progenitor differentiation. Next-generation transcriptomics sequencing showed that moringin treatment increased the expression of genes involved in neuron cortical development and in particular in neuron belonging to upper and deep cortical layers. Moreover, moringin treatment upregulated genes involved in osteogenesis and adipogenesis although with a lower fold change compared to upregulated genes involved in neuronal differentiation. Finally, moringin did not induce the expression of oncogenes resulting in a safe treatment.
Collapse
Affiliation(s)
- Letizia Romeo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Francesca Diomede
- Department of medical, oral and biotechnological sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Domenico Scionti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Fabrizio Lo Giudice
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Veronica Lanza Cariccio
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Renato Iori
- Consiglio per la ricerca in agricoltura e l'analisi dell'economia agraria, Centro di ricerca Agricoltura e Ambiente (CREA-AA), Via di Corticella 133, 40128, Bologna, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Oriana Trubiani
- Department of medical, oral and biotechnological sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy.
| |
Collapse
|
27
|
Diomede F, Gugliandolo A, Cardelli P, Merciaro I, Ettorre V, Traini T, Bedini R, Scionti D, Bramanti A, Nanci A, Caputi S, Fontana A, Mazzon E, Trubiani O. Three-dimensional printed PLA scaffold and human gingival stem cell-derived extracellular vesicles: a new tool for bone defect repair. Stem Cell Res Ther 2018; 9:104. [PMID: 29653587 PMCID: PMC5899396 DOI: 10.1186/s13287-018-0850-0] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/27/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The role of bone tissue engineering in the field of regenerative medicine has been a main research topic over the past few years. There has been much interest in the use of three-dimensional (3D) engineered scaffolds (PLA) complexed with human gingival mesenchymal stem cells (hGMSCs) as a new therapeutic strategy to improve bone tissue regeneration. These devices can mimic a more favorable endogenous microenvironment for cells in vivo by providing 3D substrates which are able to support cell survival, proliferation and differentiation. The present study evaluated the in vitro and in vivo capability of bone defect regeneration of 3D PLA, hGMSCs, extracellular vesicles (EVs), or polyethyleneimine (PEI)-engineered EVs (PEI-EVs) in the following experimental groups: 3D-PLA, 3D-PLA + hGMSCs, 3D-PLA + EVs, 3D-PLA + EVs + hGMSCs, 3D-PLA + PEI-EVs, 3D-PLA + PEI-EVs + hGMSCs. METHODS The structural parameters of the scaffold were evaluated using both scanning electron microscopy and nondestructive microcomputed tomography. Nanotopographic surface features were investigated by means of atomic force microscopy. Scaffolds showed a statistically significant mass loss along the 112-day evaluation. RESULTS Our in vitro results revealed that both 3D-PLA + EVs + hGMSCs and 3D-PLA + PEI-EVs + hGMSCs showed no cytotoxicity. However, 3D-PLA + PEI-EVs + hGMSCs exhibited greater osteogenic inductivity as revealed by morphological evaluation and transcriptomic analysis performed by next-generation sequencing (NGS). In addition, in vivo results showed that 3D-PLA + PEI-EVs + hGMSCs and 3D-PLA + PEI-EVs scaffolds implanted in rats subjected to cortical calvaria bone tissue damage were able to improve bone healing by showing better osteogenic properties. These results were supported also by computed tomography evaluation that revealed the repair of bone calvaria damage. CONCLUSION The re-establishing of the integrity of the bone lesions could be a promising strategy in the treatment of accidental or surgery trauma, especially for cranial bones.
Collapse
Affiliation(s)
- Francesca Diomede
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | | | - Paolo Cardelli
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Ilaria Merciaro
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Valeria Ettorre
- Department of Pharmacy, University "G. d'Annunzio", Chieti, Italy
| | - Tonino Traini
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Rossella Bedini
- National Centre of Innovative Technologies in Public Health, Italian National Institute of Health, Rome, Italy
| | | | - Alessia Bramanti
- IRCCS Centro Neurolesi "Bonino Pulejo", Messina, Italy.,Institute of Applied Science and Intelligent Systems "ISASI Eduardo Caianiello", CNR, Messina, Italy
| | - Antonio Nanci
- Laboratory for the study of Calcified Tissues and Biomaterials, Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, Québec, Canada
| | - Sergio Caputi
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | | | | | - Oriana Trubiani
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Via dei Vestini, 66100, Chieti, Italy.
| |
Collapse
|
28
|
Adipose mesenchymal stem cells from osteoporotic donors preserve functionality and modulate systemic inflammatory microenvironment in osteoporotic cytotherapy. Sci Rep 2018; 8:5215. [PMID: 29581449 PMCID: PMC5980002 DOI: 10.1038/s41598-018-23098-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 03/05/2018] [Indexed: 12/24/2022] Open
Abstract
Maintenance of bone homeostasis against diseased microenvironments remains as a major challenge. Recently, mesenchymal stem cells (MSCs) have been unravelled as potent microenvironmental modulators, the systemic infusion of which in cytotherapy can prevent or rescue extensive bone loss via anti-inflammation. However, MSCs also accept microenvironmental regulations; particularly, MSCs from bone marrow (BMMSCs) are prone to pathological microenvironmental factors of bone. In this study, we discovered that BMMSCs from osteoporotic donors of ovariectomized (OVX) mice lost their anti-inflammatory capability and failed to prevent bone loss when infused back into OVX recipients. Nevertheless, MSCs from adipose tissues (ADMSCs) preserved their anti-inflammatory capacity, despite diseased microenvironments of OVX donors, and continued to show protective effects on bone in OVX recipients. In the cellular level, the anti-inflammatory superiority of osteoporotic donor-derived ADMSCs over BMMSCs existed in their distinctive capability to induce T-cell apoptosis, which was molecularly attributed to retained expression levels of critical immunomodulatory genes. Furthermore, these functional discrepancies of BMMSCs and ADMSCs were due to differential stemness, energy metabolism and anti-oxidative defence system, underlying general disparity in their cellular states. Collectively, our findings optimize osteoporotic cytotherapy by using ADMSCs in resistance to and in modulation of diseased microenvironments.
Collapse
|
29
|
Robert AW, Angulski ABB, Spangenberg L, Shigunov P, Pereira IT, Bettes PSL, Naya H, Correa A, Dallagiovanna B, Stimamiglio MA. Gene expression analysis of human adipose tissue-derived stem cells during the initial steps of in vitro osteogenesis. Sci Rep 2018; 8:4739. [PMID: 29549281 PMCID: PMC5856793 DOI: 10.1038/s41598-018-22991-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/06/2018] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely studied with regard to their potential use in cell therapy protocols and regenerative medicine. However, a better comprehension about the factors and molecular mechanisms driving cell differentiation is now mandatory to improve our chance to manipulate MSC behavior and to benefit future applications. In this work, we aimed to study gene regulatory networks at an early step of osteogenic differentiation. Therefore, we analyzed both the total mRNA and the mRNA fraction associated with polysomes on human adipose tissue-derived stem cells (hASCs) at 24 h of osteogenesis induction. The RNA-seq results evidenced that hASC fate is not compromised with osteogenesis at this time and that 21 days of continuous cell culture stimuli are necessary for full osteogenic differentiation of hASCs. Furthermore, early stages of osteogenesis induction involved gene regulation that was linked to the management of cell behavior in culture, such as the control of cell adhesion and proliferation. In conclusion, although discrete initial gene regulation related to osteogenesis occur, the first 24 h of induction is not sufficient to trigger and drive in vitro osteogenic differentiation of hASCs.
Collapse
Affiliation(s)
- Anny Waloski Robert
- Instituto Carlos Chagas, Fiocruz-Paraná. Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Addeli Bez Batti Angulski
- Instituto Carlos Chagas, Fiocruz-Paraná. Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Lucia Spangenberg
- Unidad de Bioinformática, Institut Pasteur Montevideo. Mataojo 2020, Montevideo, 11400, Uruguay
| | - Patrícia Shigunov
- Instituto Carlos Chagas, Fiocruz-Paraná. Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Isabela Tiemy Pereira
- Instituto Carlos Chagas, Fiocruz-Paraná. Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | | | - Hugo Naya
- Unidad de Bioinformática, Institut Pasteur Montevideo. Mataojo 2020, Montevideo, 11400, Uruguay
| | - Alejandro Correa
- Instituto Carlos Chagas, Fiocruz-Paraná. Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Bruno Dallagiovanna
- Instituto Carlos Chagas, Fiocruz-Paraná. Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Marco Augusto Stimamiglio
- Instituto Carlos Chagas, Fiocruz-Paraná. Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil.
| |
Collapse
|
30
|
Castellano-Castillo D, Moreno-Indias I, Fernández-García JC, Alcaide-Torres J, Moreno-Santos I, Ocaña L, Gluckman E, Tinahones F, Queipo-Ortuño MI, Cardona F. Adipose Tissue LPL Methylation is Associated with Triglyceride Concentrations in the Metabolic Syndrome. Clin Chem 2017; 64:210-218. [PMID: 29046332 DOI: 10.1373/clinchem.2017.277921] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 09/12/2017] [Indexed: 02/03/2023]
Abstract
BACKGROUND DNA methylation is one of the epigenetic mechanisms that regulate gene expression. DNA methylation may be modified by environmental and nutritional factors. Thus, epigenetics could potentially provide a mechanism to explain the etiology of metabolic disorders, such as metabolic syndrome (MetS). The aim of this study was to analyze the level of DNA methylation of several lipoprotein lipase (LPL)-promoter-CpG dinucleotides in a CpG island region and relate this to the gene and protein expression levels in human visceral adipose tissue (VAT) from individuals with and without MetS. METHODS VAT samples were collected from laparoscopic surgical patients without and with MetS, and levels of LPL mRNA, LPL protein, and LPL DNA methylation were measured by qPCR, western blot, and pyrosequencing. Biochemical and anthropometric variables were analyzed. Individuals included in a subset underwent a dietary fat challenge test, and levels of postprandial triglycerides were determined. RESULTS We found higher levels of DNA methylation in MetS patients but lower gene expression and protein levels. There was a negative association between LPL methylation and LPL gene expression. We found a positive association between LPL methylation status and abnormalities of the metabolic profile and basal and postprandial triglycerides, whereas LPL gene expression was negatively associated with these abnormalities. CONCLUSIONS We demonstrate that LPL methylation may be influenced by the degree of metabolic disturbances and could be involved in triglyceride metabolism, promoting hypertriglyceridemia and subsequent associated disorders, such as MetS.
Collapse
Affiliation(s)
- Daniel Castellano-Castillo
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Madrid, Spain
| | - Isabel Moreno-Indias
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Madrid, Spain
| | - José Carlos Fernández-García
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Madrid, Spain
| | - Juan Alcaide-Torres
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Madrid, Spain
| | - Inmaculada Moreno-Santos
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, Universidad de Málaga, Red de Investigación Cardiovascular (RIC), Málaga, Spain
| | - Luis Ocaña
- Unidad de Cirugía Metabólica, Hospital Clínico Virgen de la Victoria, Málaga, Spain
| | - Enrique Gluckman
- Unidad de Cirugía Metabólica, Hospital Clínico Virgen de la Victoria, Málaga, Spain
| | - Francisco Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Madrid, Spain
| | - María Isabel Queipo-Ortuño
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain; .,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Madrid, Spain
| | - Fernando Cardona
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Madrid, Spain
| |
Collapse
|
31
|
Arrizabalaga JH, Nollert MU. Properties of porcine adipose-derived stem cells and their applications in preclinical models. Adipocyte 2017; 6:217-223. [PMID: 28410000 DOI: 10.1080/21623945.2017.1312040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adipose-derived stem cells represent a reliable adult stem cell source thanks to their abundance, straightforward isolation, and broad differentiation abilities. Consequently, human adipose-derived stem cells (hASCs) have been used in vitro for several innovative cellular therapy and regenerative medicine applications. However, the translation of a novel technology from the laboratory to the clinic requires first to evaluate its safety, feasibility, and potential efficacy through preclinical studies in animals. The anatomy and physiology of pigs and humans are very similar, establishing pigs as an attractive and popular large animal model for preclinical studies. Knowledge of the properties of porcine adipose-derived stem cells (pASCs) used in preclinical studies is critical for their success. While hASCs have been extensively studied this past decade, only a handful of reports relate to pASCs. The aim of this concise review is to summarize the current findings about the isolation of pASCs, their culture, proliferation, and immunophenotype. The differentiation abilities of pASCs and their applications in porcine preclinical models will also be reported.
Collapse
Affiliation(s)
| | - Matthias U. Nollert
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- School of Chemical, Biological & Materials Engineering, University of Oklahoma, Norman, OK, USA
| |
Collapse
|
32
|
Pedron S, Pritchard AM, Vincil GA, Andrade B, Zimmerman SC, Harley BA. Patterning Three-Dimensional Hydrogel Microenvironments Using Hyperbranched Polyglycerols for Independent Control of Mesh Size and Stiffness. Biomacromolecules 2017; 18:1393-1400. [PMID: 28245360 PMCID: PMC5444810 DOI: 10.1021/acs.biomac.7b00118] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The extracellular matrix is an environment rich with structural, mechanical, and molecular signals that can impact cell biology. Traditional approaches in hydrogel biomaterial design often rely on modifying the concentration of cross-linking groups to adjust mechanical properties. However, this strategy provides limited capacity to control additional important parameters in 3D cell culture such as microstructure and molecular diffusivity. Here we describe the use of multifunctional hyperbranched polyglycerols (HPGs) to manipulate the mechanical properties of polyethylene glycol (PEG) hydrogels while not altering biomolecule diffusion. This strategy also provides the ability to separately regulate spatial and temporal distribution of biomolecules tethered within the hydrogel. The functionalized HPGs used here can also react through a copper-free click chemistry, allowing for the encapsulation of cells and covalently tethered biomolecules within the hydrogel. Because of the hyperbranched architecture and unique properties of HPGs, their addition into PEG hydrogels affords opportunities to locally alter hydrogel cross-linking density with minimal effects on global network architecture. Additionally, photocoupling chemistry allows micropatterning of bioactive cues within the three-dimensional gel structure. This approach therefore enables us to tailor mechanical and diffusive properties independently while further allowing for local modulation of biomolecular cues to create increasingly complex cell culture microenvironments.
Collapse
Affiliation(s)
- Sara Pedron
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA
| | - Amanda M. Pritchard
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA
| | - Gretchen A. Vincil
- Department of Chemistry, University of Illinois at Urbana-Champaign, 505 South Mathews Avenue, Urbana, IL 61801, USA
| | - Brenda Andrade
- Department of Chemistry, University of Illinois at Urbana-Champaign, 505 South Mathews Avenue, Urbana, IL 61801, USA
| | - Steven C. Zimmerman
- Department of Chemistry, University of Illinois at Urbana-Champaign, 505 South Mathews Avenue, Urbana, IL 61801, USA
| | - Brendan A.C. Harley
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA
- Dept. of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Lab., 600 S. Mathews Avenue, Urbana, IL 61801, USA
| |
Collapse
|
33
|
Rubessa M, Polkoff K, Bionaz M, Monaco E, Milner DJ, Holllister SJ, Goldwasser MS, Wheeler MB. Use of Pig as a Model for Mesenchymal Stem Cell Therapies for Bone Regeneration. Anim Biotechnol 2017; 28:275-287. [PMID: 28267421 DOI: 10.1080/10495398.2017.1279169] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bone is a plastic tissue with a large healing capability. However, extensive bone loss due to disease or trauma requires extreme therapy such as bone grafting or tissue-engineering applications. Presently, bone grafting is the gold standard for bone repair, but presents serious limitations including donor site morbidity, rejection, and limited tissue regeneration. The use of stem cells appears to be a means to overcome such limitations. Bone marrow mesenchymal stem cells (BMSC) have been the choice thus far for stem cell therapy for bone regeneration. However, adipose-derived stem cells (ASC) have similar immunophenotype, morphology, multilineage potential, and transcriptome compared to BMSC, and both types have demonstrated extensive osteogenic capacity both in vitro and in vivo in several species. The use of scaffolds in combination with stem cells and growth factors provides a valuable tool for guided bone regeneration, especially for complex anatomic defects. Before translation to human medicine, regenerative strategies must be developed in animal models to improve effectiveness and efficiency. The pig presents as a useful model due to similar macro- and microanatomy and favorable logistics of use. This review examines data that provides strong support for the clinical translation of the pig model for bone regeneration.
Collapse
Key Words
- ASC, adipose-derived stem cells
- BMP, bone morphogenetic protein
- BMSC, bone marrow mesenchymal stem cells
- Bone
- DEG, differentially expressed genes
- FDR, false-discovery rate
- HA, hydroxyapatite
- HA/TCP, hydroxyapatite/tricalcium phosphate
- MRI, magnetic resonance imaging
- MSC, mesenchymal stem cells
- ONFH, osteonecrosis of the femoral head
- PCL, Poly (ϵ-caprolactone)
- PEG, polyethylene glycol
- PLGA, polylactic-coglycolic acid
- TCP, beta tri-calcium phosphate
- USSC, unrestricted somatic stem cell
- scaffolds
- stem cells
- swine
- tissue engineering
Collapse
Affiliation(s)
- Marcello Rubessa
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | - Kathryn Polkoff
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | | | - Elisa Monaco
- b Oregon State University , Corvallis , Oregon , USA
| | - Derek J Milner
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | | | - Michael S Goldwasser
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA.,d New Hanover Regional Medical Center , Wilmington , North Carolina , USA
| | - Matthew B Wheeler
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| |
Collapse
|
34
|
Wang JY, Chen WM, Wen CS, Hung SC, Chen PW, Chiu JH. Du-Huo-Ji-Sheng-Tang and its active component Ligusticum chuanxiong promote osteogenic differentiation and decrease the aging process of human mesenchymal stem cells. JOURNAL OF ETHNOPHARMACOLOGY 2017; 198:64-72. [PMID: 28040510 DOI: 10.1016/j.jep.2016.12.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 09/01/2016] [Accepted: 12/10/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Postmenopausal osteoporosis is the most common bone disease worldwide. Information concerning the effects of herbal medicines on mesenchymal cell osteogenesis and senescence remains lacking. AIM OF THIS STUDY This study was designed to investigate the effects of Du-Huo-Ji-Sheng-Tang (DHJST), a Chinese herbal medicine and its active component Ligusticum chuanxiong on osteogenic differentiation and the aging process of human mesenchymal cells (hMSCs). MATERIALS & METHODS hMSCs were used as in vitro model and osteogenesis was induced by administration of either osteogenesis inducing medium (OIM) or dexamethasone-depleted OIM (DDOIM) for 1-week or 2 weeks and the results were evaluated by measuring the formation of mineralization nodules. The effects of the compound recipe DHJST and its active component L. chuanxiong on hMSCs osteogenesis-related gene expression was determined by real-time PCR that targeted bone morphogenetic protein-2 (BMP2), RUNX2, ALP, COL-1, osteopontin (OPN), and osteocalcin (OCN). Antibodies against BMP-related signaling pathway proteins, such as BMP-2, ERK, SMAD 1/5/8, and RUNX2, were also detected at the protein level by Western blotting. Finally, the cumulative growth curve and senescence of the hMSCs were evaluated in order to assess the aging process. RESULTS L. chuanxiong increased osteogenic activity in hMSCs and up-regulated BMP-2 and RUNX2 gene expression via the activation of SMAD 1/5/8 and ERK signaling. Furthermore DHJST also showed a trend towards promoting the same effects in the same system. In the absence of dexamethasone, DHJST did activate SMAD 1/5/8 and ERK signaling and hence increased RUNX2 protein expression in hMSCs. In addition, both DHJST and L. chuanxiong delayed the hMSCs aging process by decreasing cell senescence. CONCLUSIONS We concluded that DHJST and its active component L. chuanxiong are able to promote osteogenic activity and decrease hMSCs senescence as cells age.
Collapse
Affiliation(s)
- Jir-You Wang
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC; Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
| | - Wei-Ming Chen
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Che-Sheng Wen
- Department of Orthopedics, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Shih-Chieh Hung
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Pei-Wen Chen
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Jen-Hwey Chiu
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC; Division of General Surgery, Departml;ent of Surgery, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC; Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
35
|
Skubis A, Sikora B, Zmarzły N, Wojdas E, Mazurek U. Adipose-derived stem cells: a review of osteogenesis differentiation. ACTA ACUST UNITED AC 2016. [DOI: 10.1515/fobio-2016-0004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This review article provides an overview on adipose-derived stem cells (ADSCs) for implications in bone tissue regeneration. Firstly this article focuses on mesenchymal stem cells (MSCs) which are object of interest in regenerative medicine. Stem cells have unlimited potential for self-renewal and develop into various cell types. They are used for many therapies such as bone tissue regeneration. Adipose tissue is one of the main sources of mesenchymal stem cells (MSCs). Regenerative medicine intends to differentiate ADSC along specific lineage pathways to effect repair of damaged or failing organs. For further clinical applications it is necessary to understand mechanisms involved in ADSCs proliferation and differentiation. Second part of manuscript based on osteogenesis differentiation of stem cells. Bones are highly regenerative organs but there are still many problems with therapy of large bone defects. Sometimes there is necessary to make a replacement or expansion new bone tissue. Stem cells might be a good solution for this especially ADSCs which manage differentiate into osteoblast in in vitro and in vivo conditions.
Collapse
|
36
|
Kociucka B, Flisikowska T, Mróz D, Szczerbal I. Expression of genes involved in lipid droplet formation (BSCL2, SNAP23 and COPA) during porcine in vitro adipogenesis. J Appl Genet 2016; 57:505-510. [PMID: 27108337 PMCID: PMC5061828 DOI: 10.1007/s13353-016-0350-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 04/04/2016] [Accepted: 04/15/2016] [Indexed: 11/01/2022]
Abstract
Adipogenesis is a complex process of fat cells development driven by the expression of numerous genes. Differentiation of progenitor cells into mature adipocytes is accompanied by changes in cell shape, as a result of lipid accumulation. In the present study, expression of three genes involved in lipid droplet formation (SNAP23, BSCL2 and COPA) was evaluated during porcine adipogenesis. It was found that mRNA levels of BSCL2 and SNAP23, but not COPA, increased during differentiation. Redistribution of SNAP23 protein to different cellular compartments was observed when comparing undifferentiated mesenchymal stem cells and differentiated adipocytes. The BSCL2 protein was found to be highly specific to cells with accumulated lipids, while COPA protein coated the lipid droplets. Obtained results indicated that the studied genes may be considered as candidates for fatness traits in pigs. Moreover, this study has shown that the porcine in vitro adipogenesis system provides a useful tool for the characterisation of novel genes involved in adipose tissue accumulation.
Collapse
Affiliation(s)
- Beata Kociucka
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637, Poznan, Poland
| | - Tatiana Flisikowska
- Chair of Livestock Biotechnology, Technische Universität München, München, Germany
| | - Dariusz Mróz
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637, Poznan, Poland
| | - Izabela Szczerbal
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637, Poznan, Poland.
| |
Collapse
|
37
|
Almalki SG, Agrawal DK. Key transcription factors in the differentiation of mesenchymal stem cells. Differentiation 2016; 92:41-51. [PMID: 27012163 DOI: 10.1016/j.diff.2016.02.005] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/15/2016] [Accepted: 02/25/2016] [Indexed: 11/15/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that represent a promising source for regenerative medicine. MSCs are capable of osteogenic, chondrogenic, adipogenic and myogenic differentiation. Efficacy of differentiated MSCs to regenerate cells in the injured tissues requires the ability to maintain the differentiation toward the desired cell fate. Since MSCs represent an attractive source for autologous transplantation, cellular and molecular signaling pathways and micro-environmental changes have been studied in order to understand the role of cytokines, chemokines, and transcription factors on the differentiation of MSCs. The differentiation of MSC into a mesenchymal lineage is genetically manipulated and promoted by specific transcription factors associated with a particular cell lineage. Recent studies have explored the integration of transcription factors, including Runx2, Sox9, PPARγ, MyoD, GATA4, and GATA6 in the differentiation of MSCs. Therefore, the overexpression of a single transcription factor in MSCs may promote trans-differentiation into specific cell lineage, which can be used for treatment of some diseases. In this review, we critically discussed and evaluated the role of transcription factors and related signaling pathways that affect the differentiation of MSCs toward adipocytes, chondrocytes, osteocytes, skeletal muscle cells, cardiomyocytes, and smooth muscle cells.
Collapse
Affiliation(s)
- Sami G Almalki
- Departments of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K Agrawal
- Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA.
| |
Collapse
|