1
|
Abstract
Osteopetrosis (OPT) is a rare inherited bone disease characterized by a bone resorption defect, due to osteoclast malfunction (in osteoclast-rich, oc-rich, OPT forms) or absence (in oc-poor OPT forms). This causes severe clinical abnormalities, including increased bone density, lack of bone marrow cavity, stunted growth, macrocephaly, progressive deafness, blindness, hepatosplenomegaly, and severe anemia. The oc-poor subtype of OPT is ultra-rare in humans. It is caused by mutations in either the tumor necrosis factor ligand superfamily member 11 (TNFSF11) gene, encoding RANKL (Receptor Activator of Nuclear factor-kappa B [NF-κB] Ligand) which is expressed on cells of mesenchymal origin and lymphocytes, or the TNFRSF member 11A (TNFRSF11A) gene, encoding the RANKL functional receptor RANK which is expressed on cells of myeloid lineage including osteoclasts. Clinical presentation is usually severe with onset in early infancy or in fetal life, although as more patients are reported, expressivity is variable. Phenotypic variability of RANK-deficient OPT sometimes includes hypogammaglobulinemia or radiological features of dysosteosclerosis. Disease progression is somewhat slower in RANKL-deficient OPT than in other 'malignant' subtypes of OPT. While both RANKL and RANK are essential for normal bone turnover, hematopoietic stem cell transplantation (HSCT) is the treatment of choice only for patients with the RANK-deficient form of oc-poor OPT. So far, there is no cure for RANKL-deficient OPT.
Collapse
Affiliation(s)
- Cristina Sobacchi
- CNR-IRGB, Milan Unit, via Fantoli 16/15, 20138 Milan, Italy; Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, MI, Italy.
| | - Mario Abinun
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
2
|
Zeytin IC, Alkan B, Ozdemir C, Cetinkaya DU, Okur FV. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:310-321. [PMID: 35356978 PMCID: PMC8969067 DOI: 10.1093/stcltm/szab019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/07/2021] [Indexed: 11/22/2022] Open
Abstract
Osteopetrosis is a rare inherited disease characterized by impaired osteoclast activity causing defective bone resorption and bone marrow aplasia. It is fatal in early childhood unless hematopoietic stem cell transplantation is performed. But, the transplant course is complicated with engraftment failure. Recently, osteoclasts have been described as the potential regulators of hematopoietic stem cell (HSC) niche. Here we investigated the alterations in the HSC and mesenchymal stromal cell (MSC) components of osteopetrotic niche and their interactions to mimic the stem cell dynamics/trafficking in the BM niche after HSC transplantation. Induced pluripotent stem cells were generated from peripheral blood mononuclear cells of patients with osteopetrosis carrying TCIRG1 mutation. iPSC lines were differentiated into hematopoietic and myeloid progenitors, then into osteoclasts using a step-wise protocol. We first demonstrated a shift toward monocyte-macrophages lineage regarding hematopoietic differentiation potential of osteopetrotic iPSC-derived hematopoietic progenitors (HPCs) and phenotypically normal and functionally defective osteoclast formation. The expression of the genes involved in HSC homing and maintenance (Sdf-1, Jagged-1, Kit-L, and Opn) in osteopetrotic MSCs recovered significantly after coculture with healthy HPCs. Similarly, the restoration of phenotype, impaired differentiation, and migratory potential of osteopetrotic iHPCs were observed upon interaction with healthy MSCs. Our results establish significant alterations in both MSC and HPC compartments of the osteopetrotic niche, and support the impact of functionally impaired osteoclasts in defective niche formation.
Collapse
Affiliation(s)
- Inci Cevher Zeytin
- Center for Stem Cell Research and Development PEDI-STEM, Hacettepe University, Ankara, Turkey
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Berna Alkan
- Center for Stem Cell Research and Development PEDI-STEM, Hacettepe University, Ankara, Turkey
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Cansu Ozdemir
- Center for Stem Cell Research and Development PEDI-STEM, Hacettepe University, Ankara, Turkey
| | - Duygu Uckan Cetinkaya
- Center for Stem Cell Research and Development PEDI-STEM, Hacettepe University, Ankara, Turkey
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
- Department of Pediatrics, Division of Pediatric Hematology and Bone Marrow Transplantation Unit, Faculty of Medicine, Hacettepe University, Ankara, Turkey
- Corresponding authors: Duygu Uckan Cetinkaya and Fatma Visal Okur, Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Turkey, (F.V.O.), (D.U.C.)
| | - Fatma Visal Okur
- Center for Stem Cell Research and Development PEDI-STEM, Hacettepe University, Ankara, Turkey
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
- Department of Pediatrics, Division of Pediatric Hematology and Bone Marrow Transplantation Unit, Faculty of Medicine, Hacettepe University, Ankara, Turkey
- Corresponding authors: Duygu Uckan Cetinkaya and Fatma Visal Okur, Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Turkey, (F.V.O.), (D.U.C.)
| |
Collapse
|
3
|
Güney-Esken G, Erol ÖD, Pervin B, Gürhan Sevinç G, Önder T, Bilgiç E, Korkusuz P, Günel-Özcan A, Uçkan-Çetinkaya D, Aerts-Kaya F. Development, characterization, and hematopoietic differentiation of Griscelli syndrome type 2 induced pluripotent stem cells. Stem Cell Res Ther 2021; 12:287. [PMID: 33985578 PMCID: PMC8117610 DOI: 10.1186/s13287-021-02364-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/29/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Griscelli syndrome type 2 (GS-2) is a rare, autosomal recessive immune deficiency syndrome caused by a mutation in the RAB27A gene, which results in the absence of a protein involved in vesicle trafficking and consequent loss of function of in particular cytotoxic T and NK cells. Induced pluripotent stem cells (iPSC) express genes associated with pluripotency, have the capacity for infinite expansion, and can differentiate into cells from all three germ layers. They can be induced using integrative or non-integrative systems for transfer of the Oct4, Sox2, Klf4, and cMyc (OSKM) transcription factors. To better understand the pathophysiology of GS-2 and to test novel treatment options, there is a need for an in vitro model of GS-2. METHODS Here, we generated iPSCs from 3 different GS-2 patients using lentiviral vectors. The iPSCs were characterized using flow cytometry and RT-PCR and tested for the expression of pluripotency markers. In vivo differentiation to cells from all three germlines was tested using a teratoma assay. In vitro differentiation of GS-2 iPSCs into hematopoietic stem and progenitor cells was done using Op9 feeder layers and specified media. RESULTS All GS-2 iPSC clones displayed a normal karyotype (46XX or 46XY) and were shown to express the same RAB27A gene mutation that was present in the original somatic donor cells. GS-2 iPSCs expressed SSEA1, SSEA4, TRA-1-60, TRA-1-81, and OCT4 proteins, and SOX2, NANOG, and OCT4 expression were confirmed by RT-PCR. Differentiation capacity into cells from all three germ layers was confirmed using the teratoma assay. GS-2 iPSCs showed the capacity to differentiate into cells of the hematopoietic lineage. CONCLUSIONS Using the lentiviral transfer of OSKM, we were able to generate different iPSC clones from 3 GS-2 patients. These cells can be used in future studies for the development of novel treatment options and to study the pathophysiology of GS-2 disease.
Collapse
Affiliation(s)
- Gülen Güney-Esken
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
| | - Özgür Doğuş Erol
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
| | - Burcu Pervin
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
| | - Gülben Gürhan Sevinç
- School of Medicine, Research Center for Translational Medicine, Koç University, Istanbul, Turkey
| | - Tamer Önder
- School of Medicine, Research Center for Translational Medicine, Koç University, Istanbul, Turkey
| | - Elif Bilgiç
- Faculty of Medicine, Department of Histology and Embryology, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
| | - Petek Korkusuz
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
- Faculty of Medicine, Department of Histology and Embryology, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
| | - Ayşen Günel-Özcan
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
| | - Duygu Uçkan-Çetinkaya
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Sıhhiye, 06100, Ankara, Turkey
- Faculty of Medicine, Department of Pediatrics, Division of Hematology, Hacettepe University, Ankara, Turkey
| | - Fatima Aerts-Kaya
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey.
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Sıhhiye, 06100, Ankara, Turkey.
- Laboratory Animals Research and Application Center (HUDHAM), Hacettepe University, Sıhhiye, 06100, Ankara, Turkey.
| |
Collapse
|
4
|
Sarıkaya A, Aydın G, Özyüncü Ö, Şahin E, Uçkan-Çetinkaya D, Aerts-Kaya F. Comparison of immune modulatory properties of human multipotent mesenchymal stromal cells derived from bone marrow and placenta. Biotech Histochem 2021; 97:79-89. [PMID: 33641543 DOI: 10.1080/10520295.2021.1885739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSC) can be isolated from many tissues, including bone marrow (BM) and placenta (PL). Human placenta can be obtained readily without invasive procedures. There may be differences, however, in differentiation capacity and immunomodulation by MSC isolated from BM or PL. The early pregnancy factor (heat shock protein 10; EPF/Hsp10) is a small protein that exhibits immunomodulatory properties. We compared BM- and PL-MSC, and assessed their efficacy for suppressing T-cell proliferation in vitro and the role of EPF/Hsp10 in this process. PL-MSC were collected from whole placenta after removal of the amniotic and chorionic membranes followed by serial enzymatic digestions. The PL-MSC were compared to BM-MSC, obtained from healthy donors. Differentiation capacity, cytokine secretion, expression and secretion of immunomodulatory molecules, immunophenotype and real time proliferation were assessed using cytokine arrays, ELISA assays, flow cytometry, immunohistochemical staining and western blotting. Whereas BM-MSC consisted of a homogeneous cell population with strong expression of mesenchymal markers, PL-MSC consisted of a mixed population of cells with variable CD73, CD90 and CD105 expression. PL-MSC exhibited a significantly greater proliferation rate than BM-MSC. The presence of both stem cells and more mature cells in the PL-MSC cultures resulted in decreased differentiation capacity and reduced efficacy of immune suppression in co-cultures with T-cells. Although robust intracellular expression of EPF/Hsp10 in both BM- and PL-MSC was observed, secretion of the protein in response to immune activating stimuli remained below detectable levels. Secretion of pro-inflammatory cytokines was significantly greater in BM-MSC than PL-MSC, whereas no difference was observed in the secretion of hematopoiesis supporting growth factors. Development of culture methods for isolation of pure populations of PL-MSC may improve the quality of the product and reproducibility of results.
Collapse
Affiliation(s)
- A Sarıkaya
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey.,Department of Histology and Embryology, Institute of Health Sciences, Sakarya University, Sakarya, Turkey
| | - G Aydın
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Ö Özyüncü
- Department of Obstetrics and Gynecology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - E Şahin
- Department of Histology and Embryology, Institute of Health Sciences, Sakarya University, Sakarya, Turkey.,Department of Basic Medical Sciences, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| | - D Uçkan-Çetinkaya
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey.,Bone Marrow Transplantation Unit, Division of Pediatric Hematology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - F Aerts-Kaya
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| |
Collapse
|
5
|
Lewis JW, Edwards JR, Naylor AJ, McGettrick HM. Adiponectin signalling in bone homeostasis, with age and in disease. Bone Res 2021; 9:1. [PMID: 33414405 PMCID: PMC7790832 DOI: 10.1038/s41413-020-00122-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/28/2020] [Accepted: 10/14/2020] [Indexed: 01/29/2023] Open
Abstract
Adiponectin is the most abundant circulating adipokine and is primarily involved in glucose metabolism and insulin resistance. Within the bone, osteoblasts and osteoclasts express the adiponectin receptors, however, there are conflicting reports on the effects of adiponectin on bone formation and turnover. Many studies have shown a pro-osteogenic role for adiponectin in in vivo murine models and in vitro: with increased osteoblast differentiation and activity, alongside lower levels of osteoclastogenesis. However, human studies often demonstrate an inverse relationship between adiponectin concentration and bone activity. Moreover, the presence of multiple isoforms of adiponectin and multiple receptor subtypes has the potential to lead to more complex signalling and functional consequences. As such, we still do not fully understand the importance of the adiponectin signalling pathway in regulating bone homeostasis and repair in health, with age and in disease. In this review, we explore our current understanding of adiponectin bioactivity in the bone; the significance of its different isoforms; and how adiponectin biology is altered in disease. Ultimately, furthering our understanding of adiponectin regulation of bone biology is key to developing pharmacological and non-pharmacological (lifestyle) interventions that target adiponectin signalling to boost bone growth and repair in healthy ageing, following injury or in disease.
Collapse
Affiliation(s)
- Jonathan W Lewis
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - James R Edwards
- Ageing & Regeneration Research Group, Botnar Research Centre, University of Oxford, Oxford, OX3 7LD, UK
| | - Amy J Naylor
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - Helen M McGettrick
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
6
|
Kindlin-3 mutation in mesenchymal stem cells results in enhanced chondrogenesis. Exp Cell Res 2021; 399:112456. [PMID: 33417921 DOI: 10.1016/j.yexcr.2020.112456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 11/20/2022]
Abstract
Identifying patient mutations driving skeletal development disorders has driven our understanding of bone development. Integrin adhesion deficiency disease is caused by a Kindlin-3 (fermitin family member 3) mutation, and its inactivation results in bleeding disorders and osteopenia. In this study, we uncover a role for Kindlin-3 in the differentiation of bone marrow mesenchymal stem cells (BMSCs) down the chondrogenic lineage. Kindlin-3 expression increased with chondrogenic differentiation, similar to RUNX2. BMSCs isolated from a Kindlin-3 deficient patient expressed chondrocyte markers, including SOX9, under basal conditions, which were further enhanced with chondrogenic differentiation. Rescue of integrin activation by a constitutively activated β3 integrin construct increased adhesion to multiple extracellular matrices and reduced SOX9 expression to basal levels. Growth plates from mice expressing a mutated Kindlin-3 with the integrin binding site ablated demonstrated alterations in chondrocyte maturation similar to that seen with the human Kindlin-3 deficient BMSCs. These findings suggest that Kindlin-3 expression mirrors RUNX2 during chondrogenesis.
Collapse
|
7
|
CXCR4 expression by mesenchymal stromal cells is lost after use of enzymatic dissociation agents, but preserved by use of non-enzymatic methods. Int J Hematol 2021; 113:5-9. [PMID: 33389659 DOI: 10.1007/s12185-020-03043-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 01/09/2023]
Abstract
In recent years, multipotent mesenchymal stromal cells (MSCs) have demonstrated tremendous potential for use in regenerative medicine. CXCR4, the receptor for CXCL12, is highly expressed by bone marrow (BM) MSCs and the CXCR4/CXCL12 axis has been shown to be important for migration and homing of BM-MSCs. Typically, MSCs used for clinical applications are collected after culture expansion using enzymatic methods, such as trypsin. Here, we compared different commercially available enzymatic and non-enzymatic methods for collection and dissociation of MSCs from culture plastics and their effects on CXCR4 expression by MSCs. We found that whereas non-enzymatic dissociation buffers and methods maintained CXCR4 expression, all tested enzymatic dissociation solutions dramatically decreased expression of CXCR4. We, therefore, strongly recommend the use of non-enzymatic dissociation methods, followed by filtration through a cell strainer to obtain single cell suspensions, in order to preserve maximal CXCR4 expression and optimal homing of cells.
Collapse
|
8
|
Aerts-Kaya F, Kilic E, Köse S, Aydin G, Cagnan I, Kuskonmaz B, Uckan-Cetinkaya D. G-CSF treatment of healthy pediatric donors affects their hematopoietic microenvironment through changes in bone marrow plasma cytokines and stromal cells. Cytokine 2020; 139:155407. [PMID: 33383380 DOI: 10.1016/j.cyto.2020.155407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/23/2020] [Accepted: 12/10/2020] [Indexed: 10/22/2022]
Abstract
Although G-CSF mobilized peripheral blood stem cell (PBSC) transplantation is commonly used in adults, bone marrow (BM) is still the preferred stem cell source in pediatric stem cell transplantation. Despite the fact that G-CSF is increasingly being used to enhance the hematopoietic stem/progenitor cell (HSPC) yield in BM transplantation (G-BM), the direct effects of G-CSF on the pediatric BM microenvironment have never been investigated. The BM hematopoietic niche provides the physical space where the HSPCs reside. This BM niche regulates HSPC quiescence and proliferation through direct interactions with other niche cells, including Mesenchymal Stromal Cells (MSCs). These cells have been shown to secrete a wide range of hematopoietic cytokines (CKs) and growth factors (GFs) involved in differentiation, retention and homing of hematopoietic cells. Here, we assessed changes in the BM microenvironment by measuring levels of 48 different CKs and GFs in G-BM and control BM (C-BM) plasma from pediatric donors. In addition, the effect of G-CSF on cell numbers and characteristics of HSPCs and MSCs was assessed. IL-16, SCGF-b, MIP-1b (all >1000 pg/mL) and RANTES (>10.000 pg/mL) were highly expressed in healthy donor pediatric BM plasma. Levels of IL-3, IL-18, GROa, MCP-3 (p<0.05) were increased in G-BM, whereas levels of RANTES (p<0.001) decreased after G-CSF treatment. We found a negative correlation with increasing age for IL2-Ra and LIF (p<0.05). In addition, a concomitant increase in the number of both hematopoietic and fibroblast colony forming units was observed, indicating that G-CSF affects both HSPC and MSC numbers. In conclusion, G-CSF treatment of healthy pediatric donors affects the hematopoietic BM microenvironment by expansion of HSPC and MSC numbers and modifying local CK and GF levels.
Collapse
Affiliation(s)
- Fatima Aerts-Kaya
- Hacettepe University, Graduate School of Health Sciences, Department of Stem Cell Sciences, Ankara, Turkey; Hacettepe University, Center for Stem Cell Research and Development, Ankara, Turkey
| | - Emine Kilic
- Hacettepe University, Center for Stem Cell Research and Development, Ankara, Turkey; Hemosoft IT and Training Services, Hacettepe Teknokent, Ankara, Turkey
| | - Sevil Köse
- Hacettepe University, Graduate School of Health Sciences, Department of Stem Cell Sciences, Ankara, Turkey; Hacettepe University, Center for Stem Cell Research and Development, Ankara, Turkey; Atılım University, Faculty of Medicine, Department of Medical Biology, Ankara, Turkey
| | - Gözde Aydin
- Hacettepe University, Graduate School of Health Sciences, Department of Stem Cell Sciences, Ankara, Turkey; Hacettepe University, Center for Stem Cell Research and Development, Ankara, Turkey
| | - Ilgin Cagnan
- Hacettepe University, Graduate School of Health Sciences, Department of Stem Cell Sciences, Ankara, Turkey; Hacettepe University, Center for Stem Cell Research and Development, Ankara, Turkey; Eastern Mediterranean University, Faculty of Arts and Sciences, Department of Biological Sciences, North Cyprus
| | - Baris Kuskonmaz
- Hacettepe University Medical Faculty, Department of Pediatrics, Division of Hematology-Bone Marrow Transplantation Unit, Ankara, Turkey
| | - Duygu Uckan-Cetinkaya
- Hacettepe University, Graduate School of Health Sciences, Department of Stem Cell Sciences, Ankara, Turkey; Hacettepe University, Center for Stem Cell Research and Development, Ankara, Turkey; Hacettepe University Medical Faculty, Department of Pediatrics, Division of Hematology-Bone Marrow Transplantation Unit, Ankara, Turkey.
| |
Collapse
|
9
|
Özdemir E, Emet A, Hashemihesar R, Yürüker ACS, Kılıç E, Uçkan Çetinkaya D, Turhan E. Articular Cartilage Regeneration Utilizing Decellularized Human Placental Scaffold, Mesenchymal Stem Cells and Platelet Rich Plasma. Tissue Eng Regen Med 2020; 17:901-908. [PMID: 33030679 DOI: 10.1007/s13770-020-00298-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/10/2020] [Accepted: 08/26/2020] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Articular cartilage repair has been a challenge in orthopedic practice due to the limited self-regenerative capability. Optimal treatment method for cartilage defects has not been defined. We investigated the effect of decellularized human placental (DHP) scaffold, mesenchymal stem cells (MSC) and platelet-rich plasma (PRP) on hyaline cartilage regeneration in a rat model. METHODS An osteochondral defect was created in trochlea region of the femur in all groups, bilaterally. No additional procedure was performed in control group (n = 14). Only the DHP scaffold was applied to the P group (n = 14). The DHP scaffold and 1 × 106 MSCs were applied to the PS group (n = 14). The DHP scaffold and PRP were applied to the PP group (n = 14). The DHP scaffold, 1 × 106 MSCs and PRP were applied to the PSP group (n = 14). Outcome measures at 12 weeks included Pineda histology score and qualitative histology. RESULTS The mean Pineda scores of P, PS, PP, and PSP groups were significantly better than the control group (p = 0.031, p = 0.002, p < 0.001, p < 0001, respectively). There was no statistically difference in mean Pineda scores of P, PS, PP, and PSP groups (p > 0.05). CONCLUSION In conclusion, the DHP scaffold appears to be a promising scaffold on hyaline cartilage regeneration. The augmentation of DHP scaffold with MSCs and PRP combinations did not enhance its efficacy on articular cartilage regeneration.
Collapse
Affiliation(s)
- Erdi Özdemir
- Department of Orthopedics and Traumatology, Faculty of Medicine, Hacettepe University, 06230, Ankara, Turkey.
| | - Abdülsamet Emet
- Department of Orthopedics and Traumatology, Faculty of Medicine, Hacettepe University, 06230, Ankara, Turkey
| | - Ramin Hashemihesar
- Department of Histology and Embryology, Faculty of Medicine, Istanbul Aydin University, 34295, Istanbul, Turkey
| | | | - Emine Kılıç
- Center for Stem Cell Research and Development, Hacettepe University, 06100, Ankara, Turkey
| | - Duygu Uçkan Çetinkaya
- Center for Stem Cell Research and Development, Hacettepe University, 06100, Ankara, Turkey
| | - Egemen Turhan
- Department of Orthopedics and Traumatology, Faculty of Medicine, Hacettepe University, 06230, Ankara, Turkey
| |
Collapse
|
10
|
Köse S, Aerts Kaya F, Kuşkonmaz B, Uçkan Çetinkaya D. Characterization of mesenchymal stem cells in mucolipidosis type II (I-cell disease). ACTA ACUST UNITED AC 2019; 43:171-178. [PMID: 31320815 PMCID: PMC6620033 DOI: 10.3906/biy-1902-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mucolipidosis type II (ML-II, I-cell disease) is a fatal inherited lysosomal storage disease caused by a deficiency of the enzyme N-acetylglucosamine-1-phosphotransferase. A characteristic skeletal phenotype is one of the many clinical manifestations of ML-II. Since the mechanisms underlying these skeletal defects in ML-II are not completely understood, we hypothesized that a defect in osteogenic differentiation of ML-II bone marrow mesenchymal stem cells (BM-MSCs) might be responsible for this skeletal phenotype. Here, we assessed and characterized the cellular phenotype of BM-MSCs from a ML-II patient before (BBMT) and after BM transplantation (ABMT), and we compared the results with BM-MSCs from a carrier and a healthy donor. Morphologically, we did not observe differences in ML-II BBMT and ABMT or carrier MSCs in terms of size or granularity. Osteogenic differentiation was not markedly affected by disease or carrier status. Adipogenic differentiation was increased in BBMT ML-II MSCs, but chondrogenic differentiation was decreased in both BBMT and ABMT ML-II MSCs. Immunophenotypically no significant differences were observed between the samples. Interestingly, the proliferative capacity of BBMT and ABMT ML-II MSCs was increased in comparison to MSCs from age-matched healthy donors. These data suggest that MSCs are not likely to cause the skeletal phenotype observed in ML-II, but they may contribute to the pathogenesis of ML-II as a result of lysosomal storage-induced pathology.
Collapse
Affiliation(s)
- Sevil Köse
- Department of Medical Biology and Genetics, Faculty of Medicine, Atılım University, Ankara, Turkey.,Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Turkey
| | - Fatima Aerts Kaya
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Turkey.,Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Barış Kuşkonmaz
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Turkey.,BMT Unit, Department of Pediatric Hematology, Hacettepe University Children's Hospital, Ankara, Turkey
| | - Duygu Uçkan Çetinkaya
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Turkey.,Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey.,BMT Unit, Department of Pediatric Hematology, Hacettepe University Children's Hospital, Ankara, Turkey
| |
Collapse
|
11
|
Sevim H, Kocaefe YÇ, Onur MA, Uçkan-Çetinkaya D, Gürpınar ÖA. Bone marrow derived mesenchymal stem cells ameliorate inflammatory response in an in vitro model of familial hemophagocytic lymphohistiocytosis 2. Stem Cell Res Ther 2018; 9:198. [PMID: 30021624 PMCID: PMC6052587 DOI: 10.1186/s13287-018-0941-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/31/2018] [Accepted: 06/25/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Familial hemophagocytic lymphohistiocytosis 2 (FHL2) is the most common familial type of hemophagocytic lymphohistiocytosis with immune dysregulation. FHL2 patients have mutations in the perforin gene which cause overactivation and proliferation of cytotoxic T lymphocytes and natural killer cells. Perforin is the key component of the cytolytic granule response function of cytotoxic T lymphocytes and natural killer cells. Perforin dysfunction causes a cytotoxic immune deficiency with a clinical outcome of uncontrolled and continuous immune stimulation response. This excessive stimulation leads to continuous systemic inflammation and, ultimately, multiorgan failure. Radical therapy is hematopoietic stem cell transplantation which is limited by the availability of a donor. Exacerbations of inflammatory attacks require a palliative immunosuppressive regimen. There is a need for an alternative or adjuvant therapy to maintain these patients when immunosuppression is ineffective or a donor is not available. Beneficial actions of mesenchymal stem cells (MSCs) have been shown in autoimmune diseases in clinical trials and are attributed to their immune-modulatory properties. This study aimed to assess the immune-modulatory effect of MSCs in an in-vitro model of FHL2. METHODS We generated a targeted mutation in the perforin gene of NK92 cells to create an in-vitro FLH2 model using Crispr/Cas technology. A coculture setup was employed to assess the immunomodulatory efficacy of MSCs. RESULTS Engineered NK92 clones did not show PRF1 mRNA expression and failed to secrete perforin upon phorbol myristate acetate-ionomycin stimulation, providing evidence for a valid FHL2 model. Coculture media of the engineered cells were investigated for the abundance of several cytokines. Coculture with MSCs revealed a reduction in major proinflammatory cytokines and an induction in anti-inflammatory and immunomodulatory cytokines compared to the parental NK92 cells. CONCLUSIONS This study shows the ameliorating effect of MSCs as an adjuvant immune modulator toward the therapy of FHL2 patients. MSCs are supportive therapy candidates for FHL2 patients under circumstances where prolonged immunosuppression is required to gain time before allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Handan Sevim
- Department of Biology, Faculty of Science, Hacettepe University, 06800, Ankara, Turkey.
| | - Yusuf Çetin Kocaefe
- Department of Medical Biology, Faculty of Medicine, Hacettepe University , 06100, Ankara, Turkey.,Department of Stem Cell Sciences, Institute of Health Sciences, Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Ankara, Turkey
| | - Mehmet Ali Onur
- Department of Biology, Faculty of Science, Hacettepe University, 06800, Ankara, Turkey
| | - Duygu Uçkan-Çetinkaya
- Department of Stem Cell Sciences, Institute of Health Sciences, Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Ankara, Turkey.,Pediatric Hematology, BMT Unit, Children's Hospital, Hacettepe University, 06100, Ankara, Turkey
| | - Özer Aylin Gürpınar
- Department of Biology, Faculty of Science, Hacettepe University, 06800, Ankara, Turkey
| |
Collapse
|
12
|
Fader KA, Nault R, Raehtz S, McCabe LR, Zacharewski TR. 2,3,7,8-Tetrachlorodibenzo-p-dioxin dose-dependently increases bone mass and decreases marrow adiposity in juvenile mice. Toxicol Appl Pharmacol 2018; 348:85-98. [PMID: 29673856 PMCID: PMC5984050 DOI: 10.1016/j.taap.2018.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/03/2018] [Accepted: 04/11/2018] [Indexed: 12/20/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and other aryl hydrocarbon receptor (AhR) agonists have been shown to regulate bone development and remodeling in a species-, ligand-, and age-specific manner, however the underlying mechanisms remain poorly understood. In this study, we characterized the effect of 0.01-30 μg/kg TCDD on the femoral morphology of male and female juvenile mice orally gavaged every 4 days for 28 days and used RNA-Seq to investigate gene expression changes associated with the resultant phenotype. Micro-computed tomography revealed that TCDD dose-dependently increased trabecular bone volume fraction (BVF) 2.9- and 3.3-fold in male and female femurs, respectively. Decreased serum tartrate-resistant acid phosphatase (TRAP) levels, combined with a reduced osteoclast surface to bone surface ratio and repression of femoral proteases (cathepsin K, matrix metallopeptidase 13), suggests that TCDD impaired bone resorption. Increased osteoblast counts at the trabecular bone surface were consistent with a reciprocal reduction in the number of bone marrow adipocytes, suggesting AhR activation may direct mesenchymal stem cell differentiation towards osteoblasts rather than adipocytes. Notably, femoral expression of transmembrane glycoprotein NMB (Gpnmb; osteoactivin), a positive regulator of osteoblast differentiation and mineralization, was dose-dependently induced up to 18.8-fold by TCDD. Moreover, increased serum levels of 1,25-dihydroxyvitamin D3 were in accordance with the renal induction of 1α-hydroxylase Cyp27b1 and may contribute to impaired bone resorption. Collectively, the data suggest AhR activation tipped the bone remodeling balance towards bone formation, resulting in increased bone mass with reduced marrow adiposity.
Collapse
Affiliation(s)
- Kelly A Fader
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Rance Nault
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Sandi Raehtz
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States
| | - Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States; Department of Radiology, Michigan State University, East Lansing, MI 48824, United States
| | - Timothy R Zacharewski
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
13
|
Total body irradiation tremendously impair the proliferation, differentiation and chromosomal integrity of bone marrow-derived mesenchymal stromal stem cells. Ann Hematol 2018; 97:697-707. [PMID: 29349655 DOI: 10.1007/s00277-018-3231-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 01/01/2018] [Indexed: 01/05/2023]
Abstract
Total body irradiation (TBI) is frequently used in hematopoietic stem cell transplantation (HSCT) and is associated with many complications due to radiation injury to the normal cells, including normal stem cells. Nevertheless, the effects of TBI on the mesenchymal stromal stem cell (MSC) are not fully understood. Bone marrow-derived MSCs (BM-MSCs) isolated from normal adults were irradiated with 200 cGy twice daily for consecutive 3 days, a regimen identical to that used in TBI-conditioning HSCT. The characteristics, differentiation potential, cytogenetics, hematopoiesis-supporting function, and carcinogenicity of the irradiated BM-MSCs were then compared to the non-irradiated control. The irradiated and non-irradiated MSCs shared similar morphology, phenotype, and hematopoiesis-supporting function. However, irradiated MSCs showed much lower proliferative and differentiative potential. Irradiation also induced clonal cytogenetic abnormalities of MSCs. Nevertheless, the carcinogenicity of irradiated MSCs is low in vitro and in vivo. In parallel with the ex vivo irradiation experiments, decreased proliferative and differentiative abilities and clonal cytogenetic abnormalities can also be found in MSCs isolated from transplant recipients who had received TBI-based conditioning previously. Thus, TBI used in HSCT drastically injury MSCs and may contribute to the development of some long-term complications associated with clonal cytogenetic abnormality and poor adipogenesis and osteogenesis after TBI.
Collapse
|
14
|
Schena F, Menale C, Caci E, Diomede L, Palagano E, Recordati C, Sandri M, Tampieri A, Bortolomai I, Capo V, Pastorino C, Bertoni A, Gattorno M, Martini A, Villa A, Traggiai E, Sobacchi C. Murine Rankl -/- Mesenchymal Stromal Cells Display an Osteogenic Differentiation Defect Improved by a RANKL-Expressing Lentiviral Vector. Stem Cells 2017; 35:1365-1377. [PMID: 28100034 DOI: 10.1002/stem.2574] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 12/02/2016] [Accepted: 12/26/2016] [Indexed: 01/08/2023]
Abstract
Autosomal recessive osteopetrosis (ARO) is a severe bone disease characterized by increased bone density due to impairment in osteoclast resorptive function or differentiation. Hematopoietic stem cell transplantation is the only available treatment; however, this therapy is not effective in RANKL-dependent ARO, since in bone this gene is mainly expressed by cells of mesenchymal origin. Of note, whether lack of RANKL production might cause a defect also in the bone marrow (BM) stromal compartment, possibly contributing to the pathology, is unknown. To verify this possibility, we generated and characterized BM mesenchymal stromal cell (BM-MSC) lines from wild type and Rankl-/- mice, and found that Rankl-/- BM-MSCs displayed reduced clonogenicity and osteogenic capacity. The differentiation defect was significantly improved by lentiviral transduction of Rankl-/- BM-MSCs with a vector stably expressing human soluble RANKL (hsRANKL). Expression of Rankl receptor, Rank, on the cytoplasmic membrane of BM-MSCs pointed to the existence of an autocrine loop possibly activated by the secreted cytokine. Based on the close resemblance of RANKL-defective osteopetrosis in humans and mice, we expect that our results are also relevant for RANKL-dependent ARO patients. Data obtained in vitro after transduction with a lentiviral vector expressing hsRANKL would suggest that restoration of RANKL production might not only rescue the defective osteoclastogenesis of this ARO form, but also improve a less obvious defect in the osteoblast lineage, thus possibly achieving higher benefit for the patients, when the approach is translated to clinics. Stem Cells 2017;35:1365-1377.
Collapse
Affiliation(s)
- Francesca Schena
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy
| | - Ciro Menale
- Milan Unit, CNR-IRGB, Milan, Italy.,Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Emanuela Caci
- Molecular Genetics Laboratory G. Gaslini Children's Hospital, Genova, Italy
| | - Lorenzo Diomede
- Milan Unit, CNR-IRGB, Milan, Italy.,Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Eleonora Palagano
- Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Camilla Recordati
- Mouse and Animal Pathology Laboratory, Fondazione Filarete, Milano, Italy
| | - Monica Sandri
- ISTEC-CNR, Institute of Science & Technology for Ceramics, National Research Council of Italy, Faenza, Italy
| | - Anna Tampieri
- ISTEC-CNR, Institute of Science & Technology for Ceramics, National Research Council of Italy, Faenza, Italy
| | - Ileana Bortolomai
- Milan Unit, CNR-IRGB, Milan, Italy.,San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Pastorino
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy
| | - Arinna Bertoni
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Marco Gattorno
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy
| | - Alberto Martini
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy
| | - Anna Villa
- Milan Unit, CNR-IRGB, Milan, Italy.,Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Elisabetta Traggiai
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy.,Novartis Institute for Biomedical Research, Klybeckstrasse, Basel, Switzerland
| | - Cristina Sobacchi
- Milan Unit, CNR-IRGB, Milan, Italy.,Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy
| |
Collapse
|
15
|
Ghali O, Al Rassy N, Hardouin P, Chauveau C. Increased Bone Marrow Adiposity in a Context of Energy Deficit: The Tip of the Iceberg? Front Endocrinol (Lausanne) 2016; 7:125. [PMID: 27695438 PMCID: PMC5025430 DOI: 10.3389/fendo.2016.00125] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/30/2016] [Indexed: 12/17/2022] Open
Abstract
Elevated bone marrow adiposity (BMA) is defined as an increase in the proportion of the bone marrow (BM) cavity volume occupied by adipocytes. This can be caused by an increase in the size and/or number of adipocytes. BMA increases with age in a bone-site-specific manner. This increase may be linked to certain pathophysiological situations. Osteoporosis or compromised bone quality is frequently associated with high BMA. The involvement of BM adipocytes in bone loss may be due to commitment of mesenchymal stem cells to the adipogenic pathway rather than the osteogenic pathway. However, adipocytes may also act on their microenvironment by secreting factors with harmful effects for the bone health. Here, we review evidence that in a context of energy deficit (such as anorexia nervosa (AN) and restriction rodent models) bone alterations can occur in the absence of an increase in BMA. In severe cases, bone alterations are even associated with gelatinous BM transformation. The relationship between BMA and energy deficit and the potential regulators of this adiposity in this context are also discussed. On the basis of clinical studies and preliminary results on animal model, we propose that competition between differentiation into osteoblasts and differentiation into adipocytes might trigger bone loss at least in moderate-to-severe AN and in some calorie restriction models. Finally, some of the main questions resulting from this hypothesis are discussed.
Collapse
Affiliation(s)
- Olfa Ghali
- Laboratoire de Physiopathologie des Maladies Osseuses Inflammatoires, Université de Lille, Boulogne-sur-Mer, France
- Laboratoire de Physiopathologie des Maladies Osseuses Inflammatoires, Université du Littoral Côte d’Opale, Boulogne-sur-Mer, France
| | - Nathalie Al Rassy
- Laboratoire de Physiopathologie des Maladies Osseuses Inflammatoires, Université de Lille, Boulogne-sur-Mer, France
- Laboratoire de Physiopathologie des Maladies Osseuses Inflammatoires, Université du Littoral Côte d’Opale, Boulogne-sur-Mer, France
| | - Pierre Hardouin
- Laboratoire de Physiopathologie des Maladies Osseuses Inflammatoires, Université de Lille, Boulogne-sur-Mer, France
- Laboratoire de Physiopathologie des Maladies Osseuses Inflammatoires, Université du Littoral Côte d’Opale, Boulogne-sur-Mer, France
| | - Christophe Chauveau
- Laboratoire de Physiopathologie des Maladies Osseuses Inflammatoires, Université de Lille, Boulogne-sur-Mer, France
- Laboratoire de Physiopathologie des Maladies Osseuses Inflammatoires, Université du Littoral Côte d’Opale, Boulogne-sur-Mer, France
- *Correspondence: Christophe Chauveau,
| |
Collapse
|
16
|
Sadie-Van Gijsen H, Hough FS, Ferris WF. Determinants of bone marrow adiposity: the modulation of peroxisome proliferator-activated receptor-γ2 activity as a central mechanism. Bone 2013; 56:255-65. [PMID: 23800517 DOI: 10.1016/j.bone.2013.06.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/04/2013] [Accepted: 06/12/2013] [Indexed: 12/23/2022]
Abstract
Although the presence of adipocytes in the bone marrow is a normal physiological phenomenon, the role of these cells in bone homeostasis and during pathological states has not yet been fully delineated. As osteoblasts and adipocytes originate from a common progenitor, with an inverse relationship existing between osteoblastogenesis and adipogenesis, bone marrow adiposity often negatively correlates with osteoblast number and bone mineral density. Bone adiposity can be affected by several physiological and pathophysiological factors, with abnormal, elevated marrow fat resulting in a pathological state. This review focuses on the regulation of bone adiposity by physiological factors, including aging, mechanical loading and growth factor expression, as well as the pathophysiological factors, including diseases such as anorexia nervosa and dyslipidemia, and pharmacological agents such as thiazolidinediones and statins. Although these factors regulate bone marrow adiposity via a plethora of different intracellular signaling pathways, these diverse pathways often converge on the modulation of the expression and/or activity of the pro-adipogenic transcription factor peroxisome proliferator-activated receptor (PPAR)-γ2, suggesting that any factor that affects PPAR-γ2 may have an impact on the fat content of bone.
Collapse
Affiliation(s)
- H Sadie-Van Gijsen
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Francie van Zijl Drive, Tygerberg 7505, South Africa.
| | | | | |
Collapse
|
17
|
Tayman C, Uckan D, Kilic E, Ulus AT, Tonbul A, Murat Hirfanoglu I, Helvacioglu F, Haltas H, Koseoglu B, Tatli MM. Mesenchymal stem cell therapy in necrotizing enterocolitis: a rat study. Pediatr Res 2011; 70:489-94. [PMID: 21772224 DOI: 10.1203/pdr.0b013e31822d7ef2] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We evaluated the potential therapeutic use of exogenous human bone marrow-derived mesenchymal stem cells (hBM-MSCs) in an experimental rat model of necrotizing enterocolitis (NEC). Thirty-six newborn Sprague-Dawley rats were randomly divided into three groups: NEC, NEC + hBM-MSC, and a control (control and control + hBM-MSC). NEC was induced by enteral formula feeding, exposure to hypoxia-hyperoxia, and cold stress. After NEC was induced, iron-labeled hBM-MSCs were administered by intraperitoneal injection. All pups were killed on the fourth day following injection, and the terminal ileum was excised for a histopathological and immunohistochemical evaluation. The pups in the NEC + hBM-MSC group showed significant weight gains and improvements in their clinical sickness scores (p < 0.01). Bowel damage severity observed in the histopathological evaluation was significantly lower in the NEC + hBM-MSC group than that in the NEC group (p = 0.012). The number of MSCs homing to the bowel was significantly higher in the NEC + hBM-MSC group than that in the control + hBM-MSC group. In conclusion, this is the first study that has evaluated the effectiveness of hBM-MSCs in a neonatal rat NEC model. MSCs reduced histopathological damage significantly.
Collapse
Affiliation(s)
- Cüneyt Tayman
- Department of Neonatology, Fatih University Faculty of Medicine, Ankara 06510, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|