1
|
Sobacchi C, Menale C, Crisafulli L, Ficara F. Role of RANKL Signaling in Bone Homeostasis. Physiology (Bethesda) 2025; 40:0. [PMID: 39255276 DOI: 10.1152/physiol.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
RANKL and its cognate receptor RANK are crucial regulators of bone metabolism in physiological as well as in pathological conditions. Here we go through the works that unveiled the paramount role of this signaling pathway. We focus on the RANKL cytokine, whose alterations are responsible for rare and common bone diseases. We describe recent insights on the regulation of RANKL expression, which provide new hints for the pharmacological regulation of this molecule. Based on the multiple functions exerted by RANKL (within and outside the bone tissue), we advise caution regarding the potential unintended consequences of its inhibition.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| | - Ciro Menale
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," Naples, Italy
| | - Laura Crisafulli
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| | - Francesca Ficara
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
2
|
Del Giudice C, Spagnuolo G, Menale C, Chou YF, Núñez Martí JM, Rengo C, Rengo S, Sauro S. Biocompatibility and osteogenic assessment of experimental fluoride-doped calcium-phosphate cements on human dental pulp stem cells. Dent Mater 2024; 40:2043-2050. [PMID: 39358191 DOI: 10.1016/j.dental.2024.09.019] [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: 08/16/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVES This study investigated the impact of some specific experimental calcium phosphate cements doped with different fluoride salts (FDCPCs) concentrations on the basal functions of human Dental Pulp Stem Cells (hDPSCs). Furthermore, this study also examined the migration, as well as the mineralisation through osteogenic differentiation. METHODS Experimental FDCPCs were formulated using different concentrations of calcium/sodium fluoride salts [(5 wt%: VS5F), (10 wt%: VS10F), (20 wt%: VS20F)]. A fluoride-free calcium phosphate (VS0F) was used as a control. The hDPSCs were assessed to evaluate their self-renewal and migration activity in the presence of eluates of the different FDCPCs. A viability assay in osteogenic conditions was carried out, along with the differentiation potential through Alkaline Phosphatase Activity (ALP), and Alizarin Red Staining (ARS). Moreover, the gene expression of specific markers (RUNX2, ALP, COL1α1, OCN, OPN, DSPP, MEPE, and DMP-1) was also evaluated. RESULTS All the tested FDCPD had no influence on cell migrations, but they caused a decrease in cell viability in osteogenic conditions when not diluted. Conversely, the eluants of VS20F showed a positive effect on stem cell differentiation. This result was corroborated through ALP activity, ARS assay. Moreover, upregulation of specific gene markers such as RUNX2, DMP-1, and DSPP was observed in hDPSCs, especially when treated with VS20F. SIGNIFICANCE The experimental FDCPC tested in this study exhibits a dose-dependent capacity to promote mineralisation in osteogenic environment. The FDCPC-VS20F seems to be the most promising experimental material suitable for developing of pulp-capping materials with osteogenic and bioactive properties.
Collapse
Affiliation(s)
- Carmela Del Giudice
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Gianrico Spagnuolo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Ciro Menale
- Department of Clinical Medicine and Surgery, the University of Naples "Federico II", 80131 Naples, Italy
| | - Yu Fu Chou
- Dental Biomaterials and Minimally Invasive Dentistry, Department of Dentistry, Faculty of Health Sciences, Cardenal Herrera-CEU University, CEU Universities, Valencia, Spain
| | - Juan Manuel Núñez Martí
- Dental Biomaterials and Minimally Invasive Dentistry, Department of Dentistry, Faculty of Health Sciences, Cardenal Herrera-CEU University, CEU Universities, Valencia, Spain
| | - Carlo Rengo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Sandro Rengo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Salvatore Sauro
- Dental Biomaterials and Minimally Invasive Dentistry, Department of Dentistry, Faculty of Health Sciences, Cardenal Herrera-CEU University, CEU Universities, Valencia, Spain.
| |
Collapse
|
3
|
Schiavone ML, Crisafulli L, Camisaschi C, De Simone G, Liberati FR, Palagano E, Rucci N, Ficara F, Sobacchi C. Rankl genetic deficiency and functional blockade undermine skeletal stem and progenitor cell differentiation. Stem Cell Res Ther 2024; 15:203. [PMID: 38971808 PMCID: PMC11227705 DOI: 10.1186/s13287-024-03803-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/16/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Skeletal Stem Cells (SSCs) are required for skeletal development, homeostasis, and repair. The perspective of their wide application in regenerative medicine approaches has supported research in this field, even though so far results in the clinic have not reached expectations, possibly due also to partial knowledge of intrinsic, potentially actionable SSC regulatory factors. Among them, the pleiotropic cytokine RANKL, with essential roles also in bone biology, is a candidate deserving deep investigation. METHODS To dissect the role of the RANKL cytokine in SSC biology, we performed ex vivo characterization of SSCs and downstream progenitors (SSPCs) in mice lacking Rankl (Rankl-/-) by means of cytofluorimetric sorting and analysis of SSC populations from different skeletal compartments, gene expression analysis, and in vitro osteogenic differentiation. In addition, we assessed the effect of the pharmacological treatment with the anti-RANKL blocking antibody Denosumab (approved for therapy in patients with pathological bone loss) on the osteogenic potential of bone marrow-derived stromal cells from human healthy subjects (hBMSCs). RESULTS We found that, regardless of the ossification type of bone, osteochondral SSCs had a higher frequency and impaired differentiation along the osteochondrogenic lineage in Rankl-/- mice as compared to wild-type. Rankl-/- mice also had increased frequency of committed osteochondrogenic and adipogenic progenitor cells deriving from perivascular SSCs. These changes were not due to the peculiar bone phenotype of increased density caused by lack of osteoclast resorption (defined osteopetrosis); indeed, they were not found in another osteopetrotic mouse model, i.e., the oc/oc mouse, and were therefore not due to osteopetrosis per se. In addition, Rankl-/- SSCs and primary osteoblasts showed reduced mineralization capacity. Of note, hBMSCs treated in vitro with Denosumab had reduced osteogenic capacity compared to control cultures. CONCLUSIONS We provide for the first time the characterization of SSPCs from mouse models of severe recessive osteopetrosis. We demonstrate that Rankl genetic deficiency in murine SSCs and functional blockade in hBMSCs reduce their osteogenic potential. Therefore, we propose that RANKL is an important regulatory factor of SSC features with translational relevance.
Collapse
Affiliation(s)
- M L Schiavone
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - L Crisafulli
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
- Institute for Genetic and Biomedical Research, Milan Unit, CNR, via Fantoli 16/15, Milan, 20138, Italy
| | - C Camisaschi
- Flow Cytometry Core, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - G De Simone
- Flow Cytometry Core, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - F R Liberati
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - E Palagano
- Institute of Biosciences and Bioresources, CNR, via Madonna Del Piano 10, Sesto Fiorentino, 50019, FI, Italy
| | - N Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, L'Aquila, 67100, Italy
| | - F Ficara
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
- Institute for Genetic and Biomedical Research, Milan Unit, CNR, via Fantoli 16/15, Milan, 20138, Italy
| | - Cristina Sobacchi
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy.
- Institute for Genetic and Biomedical Research, Milan Unit, CNR, via Fantoli 16/15, Milan, 20138, Italy.
| |
Collapse
|
4
|
Del Giudice C, Rengo C, Maglitto M, Armogida NG, Iaculli F, Rengo S, Menale C, Spagnuolo G. Cytotoxicity effects and differentiation potential of ormocer-based and nanohybrid composite resins on human dental pulp stem cells. Dent Mater 2024; 40:984-992. [PMID: 38724333 DOI: 10.1016/j.dental.2024.05.011] [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: 02/16/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE to compare conventional nanohybrid (Ceram.x Spectra) and ormocer-based (Admira fusion) dental composite resins effects on human dental pulp stem cells (hDPSCs) in terms of cytotoxicity, self-renewal, migration and osteogenic differentiation. METHODS hDPSCs were cultured in presence of different dilutions (undiluted, form 1:2 to 1:100) of CeramX (CX) and Admira fusion (AD) eluates and viability assay in standard or osteogenic conditions were performed. Samples and eluates were prepared according to ISO 10993-12. In addition, apoptosis, self-renewal and migration activity evaluations were carried out. Osteogenic differentiation potential was tested by Alkaline Phosphatase Activity, alizarin red staining and gene expression of specific markers (ALP, RUNX2, OCN, OPN and COL1α1). Statistical analysis was performed by means of a One-way analysis of variance (One-way ANOVA) followed by a Tukey's test for multiple comparison; results were presented as mean ± standard error of mean (SEM). RESULTS Admira Fusion demonstrated to be highly biocompatible and showed positive effects on hDPSCs proliferation and differentiation; on the contrary, conventional nanohybrid composite showed to be more cytotoxic and without any notable effect on stem cells differentiation. Moreover, the obtained results were further corroborated by a significant upregulation of osteogenic differentiation markers obtained in presence of ormocer-based composite resin eluate. Specifically, in AD 1:50 group expression levels of ALP, Runx2, Col1α1 were double than control (ALP, p = 0.045; Runx2, p = 0.003; Col1α1, p = 0.001) and CX 1:50 (ALP, p = 0.006; RUNX2, p = 0.029; Col1α1, p = 0.005). Moreover, in the same group, OPN and OCN resulted about 5 times more expressed as compared to control (OPN, p = 0.009; OCN, p = 0.0005) and CX 1:50 (OPN, p = 0.012; OCN, p = 0.0006). SIGNIFICANCE The less cytotoxicity obtained by AD than conventional nanohybrid composite may be attributed to a reduced monomers release in the oral environment, supporting the hypothesis of limited adverse effect and enhanced healing potential, mainly when the material is positioned in close contact with pulp tissue.
Collapse
Affiliation(s)
- C Del Giudice
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - C Rengo
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - M Maglitto
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - N G Armogida
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - F Iaculli
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy.
| | - S Rengo
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - C Menale
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - G Spagnuolo
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
5
|
Xu J, Yu L, Ye S, Ye Z, Yang L, Xu X. Oral microbiota-host interaction: the chief culprit of alveolar bone resorption. Front Immunol 2024; 15:1254516. [PMID: 38455060 PMCID: PMC10918469 DOI: 10.3389/fimmu.2024.1254516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 02/02/2024] [Indexed: 03/09/2024] Open
Abstract
There exists a bidirectional relationship between oral health and general well-being, with an imbalance in oral symbiotic flora posing a threat to overall human health. Disruptions in the commensal flora can lead to oral diseases, while systemic illnesses can also impact the oral cavity, resulting in the development of oral diseases and disorders. Porphyromonas gingivalis and Fusobacterium nucleatum, known as pathogenic bacteria associated with periodontitis, play a crucial role in linking periodontitis to accompanying systemic diseases. In periodontal tissues, these bacteria, along with their virulence factors, can excessively activate the host immune system through local diffusion, lymphatic circulation, and blood transmission. This immune response disruption contributes to an imbalance in osteoimmune mechanisms, alveolar bone resorption, and potential systemic inflammation. To restore local homeostasis, a deeper understanding of microbiota-host interactions and the immune network phenotype in local tissues is imperative. Defining the immune network phenotype in periodontal tissues offers a promising avenue for investigating the complex characteristics of oral plaque biofilms and exploring the potential relationship between periodontitis and associated systemic diseases. This review aims to provide an overview of the mechanisms underlying Porphyromonas gingivalis- and Fusobacterium nucleatum-induced alveolar bone resorption, as well as the immunophenotypes observed in host periodontal tissues during pathological conditions.
Collapse
Affiliation(s)
- Jingyu Xu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Ling Yu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Surong Ye
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zitong Ye
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Luyi Yang
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiaoxi Xu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
6
|
Giorello MB, Borzone FR, Mora MF, Padin MDR, Wernicke A, Labovsky V, Chasseing NA. RANK in cancer-associated fibroblasts: A valuable prognostic determinant for metastasis in early-stage breast cancer patients. Cancer Biomark 2024; 41:115-132. [PMID: 39240628 PMCID: PMC11492045 DOI: 10.3233/cbm-230523] [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: 01/10/2024] [Accepted: 07/19/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND The molecular system of receptor activator of nuclear factor kappa-β (RANK) and its ligand (RANKL) plays a role in a variety of physiological and pathological processes. These encompass the regulation of bone metabolism, mammary gland development, immune function, as well as their involvement and tumorigenesis. Nevertheless, limited knowledge exists regarding their function within the tumor microenvironment. METHODS AND RESULTS We explored the significance of RANK expression in cancer-associated fibroblasts (CAFs) as a prognostic biomarker in early breast cancer patients (BCPs) by immunohistochemistry. Results reveal a significant correlation between high RANK expression in CAFs and an increased risk of metastasis (p= 0.006), shorter metastasis-free survival (MFS) [p= 0.007, OR (95%CI) = 2.290 (1.259-4.156)], and lower overall survival (OS) [p= 0.004, OR (95%CI) = 2.469 (1.343-4.541)]. Upon analyzing the phenotype of CD34(-) CAFs isolated from primary tumors in BCPs, we observed co-expression of RANK with CD105 marker by immunofluorescence and flow cytometry, characteristic of mesenchymal stem/stromal cells (MSCs), suggesting the possible cellular origin. Also RANKL-RANK system increase the OCT-4, SOX-2 and DKK-1 (dickkopf 1) gene expression in CD34(-) CAFs by RT-PCR. Moreover, this system plays a crucial role in the migration of these CD34(-) CAFs. CONCLUSIONS These results support the clinical relevance of RANK in CAFs and propose its potential as a future therapeutic target in the treatment of early BCPs.
Collapse
Affiliation(s)
- María Belén Giorello
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - María Florencia Mora
- Departamento de Anatomía Patológica, Hospital Italiano, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - María del Rosario Padin
- Departamento de Anatomía Patológica, Hospital Italiano, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alejandra Wernicke
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- Departamento de Anatomía Patológica, Hospital Italiano, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Vivian Labovsky
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
7
|
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
|
8
|
Abstract
Since the receptor activator of nuclear factor-kappa B ligand (RANKL), its cognate receptor activator of nuclear factor-kappa B (RANK), and the decoy receptor osteoprotegerin (OPG) were discovered, a number of studies have uncovered the crucial role of the RANKL-RANK-OPG pathway in controlling the key aspect of bone homeostasis, the immune system, inflammation, cancer, and other systems under pathophysiological condition. These findings have expanded the understanding of the multifunctional biology of the RANKL-RANK-OPG pathway and led to the development of therapeutic potential targeting this pathway. The successful development and application of anti-RANKL antibody in treating diseases causing bone loss validates the utility of therapeutic approaches based on the modulation of this pathway. Moreover, recent studies have demonstrated the involvement of the RANKL-RANK pathway in osteoblast differentiation and bone formation, shedding light on the RANKL-RANK dual signaling in coupling bone resorption and bone formation. In this review, we will summarize the current understanding of the RANKL-RANK-OPG system in the context of the bone and the immune system as well as the impact of this pathway in disease conditions, including cancer development and metastasis.
Collapse
Affiliation(s)
- Noriko Takegahara
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Hyunsoo Kim
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Yongwon Choi
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
9
|
The polypeptide OP3-4 induced osteogenic differentiation of bone marrow mesenchymal stem cells via protein kinase B/glycogen synthase kinase 3β/β-catenin pathway and promoted mandibular defect bone regeneration. Arch Oral Biol 2021; 130:105243. [PMID: 34416564 DOI: 10.1016/j.archoralbio.2021.105243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/04/2021] [Accepted: 08/13/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES The aims of this study were to explore: (ⅰ) the effect of the polypeptide OP 3-4 on bone regeneration in vivo; (ⅱ) the effect of OP 3-4 on osteogenic differentiation of bone marrow mesenchymal stem cells in vitro; and (ⅲ) the potential mechanism of OP 3-4 in promoting osteogenic differentiation of bone marrow mesenchymal stem cells. DESIGNS 30 Wistar rats (8-week, male) were randomly divided into Control group (n = 5), Hydrogel group (n = 5), and Hydrogel loaded OP 3-4 group (n = 5). Hematoxylin and eosin staining was used to evaluate the level of bone regeneration in mandibular defect. Immunohistochemistry staining was used to evaluate the expression of alkaline phosphatase, runt-related transcription factor 2, and type Ⅰ collagen. Flow cytometry was applied to identify the phenotype of bone marrow mesenchymal stem cells. Furthermore, LY294002, the inhibitor of protein kinase B, was applied to verify the role of OP 3-4 in promoting osteogenic differentiation via protein kinase B/glycogen synthase kinase 3β/β-catenin pathway through western blot. RESULTS OP 3-4 promoted bone regeneration of rat mandibular defect. The expression of osteogenic differentiation related markers were increased after adding OP 3-4 to bone marrow mesenchymal stem cells. OP 3-4 promoted osteogenic differentiation of bone marrow mesenchymal stem cells via protein kinase B/glycogen synthase kinase 3β/β-catenin pathway. CONCLUSION OP 3-4 could promote bone regeneration of mandibular defect and improve osteogenic differentiation through protein kinase B/glycogen synthase kinase 3β/β-catenin pathway.
Collapse
|
10
|
Parente R, Possetti V, Schiavone ML, Campodoni E, Menale C, Loppini M, Doni A, Bottazzi B, Mantovani A, Sandri M, Tampieri A, Sobacchi C, Inforzato A. 3D Cocultures of Osteoblasts and Staphylococcus aureus on Biomimetic Bone Scaffolds as a Tool to Investigate the Host-Pathogen Interface in Osteomyelitis. Pathogens 2021; 10:pathogens10070837. [PMID: 34357987 PMCID: PMC8308613 DOI: 10.3390/pathogens10070837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
Osteomyelitis (OM) is an infectious disease of the bone primarily caused by the opportunistic pathogen Staphylococcus aureus (SA). This Gram-positive bacterium has evolved a number of strategies to evade the immune response and subvert bone homeostasis, yet the underlying mechanisms remain poorly understood. OM has been modeled in vitro to challenge pathogenetic hypotheses in controlled conditions, thus providing guidance and support to animal experimentation. In this regard, traditional 2D models of OM inherently lack the spatial complexity of bone architecture. Three-dimensional models of the disease overcome this limitation; however, they poorly reproduce composition and texture of the natural bone. Here, we developed a new 3D model of OM based on cocultures of SA and murine osteoblastic MC3T3-E1 cells on magnesium-doped hydroxyapatite/collagen I (MgHA/Col) scaffolds that closely recapitulate the bone extracellular matrix. In this model, matrix-dependent effects were observed in proliferation, gene transcription, protein expression, and cell–matrix interactions both of the osteoblastic cell line and of bacterium. Additionally, these had distinct metabolic and gene expression profiles, compared to conventional 2D settings, when grown on MgHA/Col scaffolds in separate monocultures. Our study points to MgHA/Col scaffolds as biocompatible and bioactive matrices and provides a novel and close-to-physiology tool to address the pathogenetic mechanisms of OM at the host–pathogen interface.
Collapse
Affiliation(s)
- Raffaella Parente
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
| | - Valentina Possetti
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
| | - Maria Lucia Schiavone
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- National Research Council-Institute for Genetic and Biomedical Research (CNR-IRGB), Milan Unit, 20089 Rozzano, Italy;
| | - Elisabetta Campodoni
- National Research Council-Institute of Science and Technology for Ceramics (CNR-ISTEC), 48018 Faenza, Italy; (E.C.); (M.S.); (A.T.)
| | - Ciro Menale
- National Research Council-Institute for Genetic and Biomedical Research (CNR-IRGB), Milan Unit, 20089 Rozzano, Italy;
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
| | - Mattia Loppini
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
| | - Andrea Doni
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
| | - Barbara Bottazzi
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
| | - Alberto Mantovani
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
- The William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK
| | - Monica Sandri
- National Research Council-Institute of Science and Technology for Ceramics (CNR-ISTEC), 48018 Faenza, Italy; (E.C.); (M.S.); (A.T.)
| | - Anna Tampieri
- National Research Council-Institute of Science and Technology for Ceramics (CNR-ISTEC), 48018 Faenza, Italy; (E.C.); (M.S.); (A.T.)
- National Research Council-Institute of Nanostructured Material (CNR-ISMN), 40129 Bologna, Italy
| | - Cristina Sobacchi
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- National Research Council-Institute for Genetic and Biomedical Research (CNR-IRGB), Milan Unit, 20089 Rozzano, Italy;
- Correspondence: (C.S.); (A.I.); Tel.: +39-028-224-5153 (C.S.); +39-028-224-5132 (A.I.)
| | - Antonio Inforzato
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
- Correspondence: (C.S.); (A.I.); Tel.: +39-028-224-5153 (C.S.); +39-028-224-5132 (A.I.)
| |
Collapse
|
11
|
Elango J, Bao B, Wu W. The hidden secrets of soluble RANKL in bone biology. Cytokine 2021; 144:155559. [PMID: 33994070 DOI: 10.1016/j.cyto.2021.155559] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/25/2022]
Abstract
The discovery of cytokine tumor necrosis factor (TNF) in the 20th century revealed numerous secrets about organ development. In particular, the functions identified for the receptor activator of nuclear factor kappa-β (NF-κβ) ligand (also known as the RANKL/osteoprotegerin ligand (OPGL) or RANK ligand/TNFSF11) in the homeostasis of skeletal structure, function and regulation were not anticipated. Empirical evidence established the receptor-ligand interaction of RANKL with RANK in osteoclast formation. Reverse signaling of RANKL triggers NF-κβ for the degradation of β-catenin to inhibit bone formation. There is also evidence that RANKL modifies the behavior of other cells in the bone microenvironment, including osteoblasts, chondrocytes, endothelial cells and lymphocytes during normal (homeostatic) and diseased (osteoimmune) states. Two forms of RANKL, i.e., soluble and membrane-bound RANKL, are produced by bone cells. Even though soluble RANKL (sRANKL) and membrane-bound RANKL (mRANKL) both stimulate osteoclast formation in vitro, their biological roles are different. mRANKL triggers osteoclastogenesis by binding to RANK through cell-cell interaction; however, sRANKL released from osteogenic cells binds to RANK without cell-cell interaction. This review attempts to hypothesize how sRANKL functions biologically in bone and explore how this hypothesis might influence future research.
Collapse
Affiliation(s)
- Jeevithan Elango
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| | - Bin Bao
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Wenhui Wu
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
12
|
Coupled OPG-Fc on Decellularized Aortic Valves by EDC/NHS Attenuates Rat MSCs Calcification In Vitro. ASAIO J 2020; 65:197-204. [PMID: 29677036 DOI: 10.1097/mat.0000000000000796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Valve calcification commonly damages natural human heart valves and tissue-engineered heart valves (TEHVs), and no ideal intervention is available in clinical practice. It is increasingly considered that osteoprotegerin (OPG) inhibits vascular calcification. Herein we aimed to explore whether free OPG-Fc fusion protein or coupled OPG-Fc on decellularized aortic valves attenuates calcification. Calcification of rat bone marrow-derived mesenchymal stromal cells (MSCs) was induced by osteogenic differentiation media, and the effects of free OPG-Fc or OPG-Fc coupled on the decellularized porcine aortic heart valve leaflet scaffolds by coupling agents 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC)/N-hydroxysuccinimide (NHS) on calcification were observed. Mineralization of the extracellular matrix, alkaline phosphatase (ALP) activity, and expression of osteoblastic markers were assessed to determine the calcification kinetics. Our results indicated that the matrix calcium content and the ALP activity, as well as the mRNA expression levels of a bone morphogenetic protein-2 (BMP-2), osteopontin (OPN), and osteocalcin (OC), of the MSCs seeded on plates with free OPG-Fc or on the OPG-Fc-coupled scaffolds decreased compared with their control MSCs without coupled OPG-Fc. The results suggest that both free and immobilized OPG-Fc on the decellularized aortic valve scaffolds by EDC/NHS can attenuate the calcification of MSCs induced by osteogenic differentiation media, implying that OPG-Fc might be a new treatment or prevention strategy for the calcification of natural human heart valves and TEHVs in the future.
Collapse
|
13
|
Physiological concentrations of denosumab enhance osteogenic differentiation in human mesenchymal stem cells of the jaw bone. Arch Oral Biol 2019; 101:23-29. [DOI: 10.1016/j.archoralbio.2019.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 02/06/2023]
|
14
|
Sobacchi C, Menale C, Villa A. The RANKL-RANK Axis: A Bone to Thymus Round Trip. Front Immunol 2019; 10:629. [PMID: 30984193 PMCID: PMC6450200 DOI: 10.3389/fimmu.2019.00629] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022] Open
Abstract
The identification of Receptor activator of nuclear factor kappa B ligand (RANKL) and its cognate receptor Receptor activator of nuclear factor kappa B (RANK) during a search for novel tumor necrosis factor receptor (TNFR) superfamily members has dramatically changed the scenario of bone biology by providing the functional and biochemical proof that RANKL signaling via RANK is the master factor for osteoclastogenesis. In parallel, two independent studies reported the identification of mouse RANKL on activated T cells and of a ligand for osteoprotegerin on a murine bone marrow-derived stromal cell line. After these seminal findings, accumulating data indicated RANKL and RANK not only as essential players for the development and activation of osteoclasts, but also for the correct differentiation of medullary thymic epithelial cells (mTECs) that act as mediators of the central tolerance process by which self-reactive T cells are eliminated while regulatory T cells are generated. In light of the RANKL-RANK multi-task function, an antibody targeting this pathway, denosumab, is now commonly used in the therapy of bone loss diseases including chronic inflammatory bone disorders and osteolytic bone metastases; furthermore, preclinical data support the therapeutic application of denosumab in the framework of a broader spectrum of tumors. Here, we discuss advances in cellular and molecular mechanisms elicited by RANKL-RANK pathway in the bone and thymus, and the extent to which its inhibition or augmentation can be translated in the clinical arena.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Milan Unit, Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Ciro Menale
- Milan Unit, Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Anna Villa
- Milan Unit, Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy.,San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
15
|
Xin X, Jiang X, Wang L, Mikael P, McCarthy MB, Chen L, Mazzocca AD, Nukavarapu S, Lichtler AC, Rowe DW. Histological Criteria that Distinguish Human and Mouse Bone Formed Within a Mouse Skeletal Repair Defect. J Histochem Cytochem 2019; 67:401-417. [PMID: 30848692 DOI: 10.1369/0022155419836436] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The effectiveness of autologous cell-based skeletal repair continues to be controversial in part because in vitro predictors of in vivo human bone formation by cultured human progenitor cells are not reliable. To assist in the development of in vivo assays of human osteoprogenitor potential, a fluorescence-based histology of nondecalcified mineralized tissue is presented that provides multiple criteria to distinguish human and host osteoblasts, osteocytes, and accumulated bone matrix in a mouse calvarial defect model. These include detection of an ubiquitously expressed red fluorescent protein reporter by the implanted human cells, antibodies specific to human bone sialoprotein and a human nuclear antigen, and expression of a bone/fibroblast restricted green fluorescent protein reporter in the host tissue. Using low passage bone marrow-derived stromal cells, robust human bone matrix formation was obtained. However, a striking feature is the lack of mouse bone marrow investment and osteoclasts within the human bone matrix. This deficiency may account for the accumulation of a disorganized human bone matrix that has not undergone extensive remodeling. These features, which would not be appreciated by traditional decalcified paraffin histology, indicate the human bone matrix is not undergoing active remodeling and thus the full differentiation potential of the implanted human cells within currently used mouse models is not being realized.
Collapse
Affiliation(s)
- Xiaonan Xin
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut
| | - Xi Jiang
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut
| | - Liping Wang
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut
| | - Paiyz Mikael
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Mary Beth McCarthy
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Li Chen
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut
| | - Augustus D Mazzocca
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Syam Nukavarapu
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut.,Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut
| | - Alexander C Lichtler
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut
| | - David W Rowe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
16
|
Penna S, Capo V, Palagano E, Sobacchi C, Villa A. One Disease, Many Genes: Implications for the Treatment of Osteopetroses. Front Endocrinol (Lausanne) 2019; 10:85. [PMID: 30837952 PMCID: PMC6389615 DOI: 10.3389/fendo.2019.00085] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 01/31/2019] [Indexed: 11/23/2022] Open
Abstract
Osteopetrosis is a condition characterized by increased bone mass due to defects in osteoclast function or formation. In the last decades, the molecular dissection of osteopetrosis has unveiled a plethora of molecular players responsible for different forms of the disease, some of which present also primary neurodegeneration that severely limits the therapy. Hematopoietic stem cell transplantation can cure the majority of them when performed in the first months of life, highlighting the relevance of an early molecular diagnosis. However, clinical management of these patients is constrained by the severity of the disease and lack of a bone marrow niche that may delay immune reconstitution. Based on osteopetrosis genetic heterogeneity and disease severity, personalized therapies are required for patients that are not candidate to bone marrow transplantation. This review briefly describes the genetics of osteopetrosis, its clinical heterogeneity, current therapy and innovative approaches undergoing preclinical evaluation.
Collapse
Affiliation(s)
- Sara Penna
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), San Raffaele Hospital, Milan, Italy
- Translational and Molecular Medicine (DIMET), University of Milano-Bicocca, Monza, Italy
| | - Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), San Raffaele Hospital, Milan, Italy
| | - Eleonora Palagano
- The National Research Council (CNR) Institute for Genetic and Biomedical Research (IRGB)- CNR-IRGB, Milan Unit, Milan, Italy
- Humanitas Research Hospital, Rozzano, Italy
| | - Cristina Sobacchi
- The National Research Council (CNR) Institute for Genetic and Biomedical Research (IRGB)- CNR-IRGB, Milan Unit, Milan, Italy
- Humanitas Research Hospital, Rozzano, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), San Raffaele Hospital, Milan, Italy
- The National Research Council (CNR) Institute for Genetic and Biomedical Research (IRGB)- CNR-IRGB, Milan Unit, Milan, Italy
- *Correspondence: Anna Villa
| |
Collapse
|
17
|
RANKL signaling in bone marrow mesenchymal stem cells negatively regulates osteoblastic bone formation. Bone Res 2018; 6:34. [PMID: 30510839 PMCID: PMC6255918 DOI: 10.1038/s41413-018-0035-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022] Open
Abstract
RANKL signaling is essential for osteoclastogenesis. Its role in osteoblastic differentiation and bone formation is unknown. Here we demonstrate that RANK is expressed at an early stage of bone marrow mesenchymal stem cells (BMSCs) during osteogenic differentiation in both mice and human and decreased rapidly. RANKL signaling inhibits osteogenesis by promoting β-catenin degradation and inhibiting its synthesis. In contrast, RANKL signaling has no significant effects on adipogenesis of BMSCs. Interestingly, conditional knockout of rank in BMSCs with Prx1-Cre mice leads to a higher bone mass and increased trabecular bone formation independent of osteoclasts. In addition, rankflox/flox: Prx1-Cre mice show resistance to ovariectomy-(OVX) induced bone loss. Thus, our results reveal that RANKL signaling regulates both osteoclasts and osteoblasts by inhibition of osteogenic differentiation of BMSCs and promotion of osteoclastogenesis. Researchers in China have shown that a gene known for breaking bones down is also involved in making new bone. Bones are constantly repaired and reshaped by cells which break down bone tissue, osteoclasts, and those which create new bone, osteoblasts. The RANKL gene is known to play an important role in osteoclast development, but Jiacan Su of the Second Military Medical University has shown that it is also important for osteoblasts. Su’s team detected high expression of RANKL and its receptor, RANK, in bone marrow stem cells, and the levels decreased as the stem cells differentiated into osteoblasts. Artificially increasing RANK expression decreased osteoblast differentiation, while reducing its expression increased osteoblast differentiation and bone mass. By showing that RANKL regulates osteoblasts as well as osteoclasts, these findings open new avenues for understanding bones development.
Collapse
|
18
|
3D Bone Biomimetic Scaffolds for Basic and Translational Studies with Mesenchymal Stem Cells. Int J Mol Sci 2018; 19:ijms19103150. [PMID: 30322134 PMCID: PMC6213614 DOI: 10.3390/ijms19103150] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/04/2018] [Accepted: 10/10/2018] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are recognized as an attractive tool owing to their self-renewal and differentiation capacity, and their ability to secrete bioactive molecules and to regulate the behavior of neighboring cells within different tissues. Accumulating evidence demonstrates that cells prefer three-dimensional (3D) to 2D culture conditions, at least because the former are closer to their natural environment. Thus, for in vitro studies and in vivo utilization, great effort is being dedicated to the optimization of MSC 3D culture systems in view of achieving the intended performance. This implies understanding cell–biomaterial interactions and manipulating the physicochemical characteristics of biomimetic scaffolds to elicit a specific cell behavior. In the bone field, biomimetic scaffolds can be used as 3D structures, where MSCs can be seeded, expanded, and then implanted in vivo for bone repair or bioactive molecules release. Actually, the union of MSCs and biomaterial has been greatly improving the field of tissue regeneration. Here, we will provide some examples of recent advances in basic as well as translational research about MSC-seeded scaffold systems. Overall, the proliferation of tools for a range of applications witnesses a fruitful collaboration among different branches of the scientific community.
Collapse
|
19
|
Menale C, Campodoni E, Palagano E, Mantero S, Erreni M, Inforzato A, Fontana E, Schena F, Van't Hof R, Sandri M, Tampieri A, Villa A, Sobacchi C. Mesenchymal Stromal Cell-Seeded Biomimetic Scaffolds as a Factory of Soluble RANKL in Rankl-Deficient Osteopetrosis. Stem Cells Transl Med 2018; 8:22-34. [PMID: 30184340 PMCID: PMC6312453 DOI: 10.1002/sctm.18-0085] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/11/2018] [Indexed: 12/27/2022] Open
Abstract
Biomimetic scaffolds are extremely versatile in terms of chemical composition and physical properties, which can be defined to accomplish specific applications. One property that can be added is the production/release of bioactive soluble factors, either directly from the biomaterial, or from cells embedded within the biomaterial. We reasoned that pursuing this strategy would be appropriate to setup a cell‐based therapy for RANKL‐deficient autosomal recessive osteopetrosis, a very rare skeletal genetic disease in which lack of the essential osteoclastogenic factor RANKL impedes osteoclast formation. The exogenously administered RANKL cytokine is effective in achieving osteoclast formation and function in vitro and in vivo, thus, we produced murine Rankl−/− mesenchymal stromal cells (MSCs) overexpressing human soluble RANKL (hsRL) following lentiviral transduction (LVhsRL). Here, we described a three‐dimensional (3D) culture system based on a magnesium‐doped hydroxyapatite/collagen I (MgHA/Col) biocompatible scaffold closely reproducing bone physicochemical properties. MgHA/Col‐seeded murine MSCs showed improved properties, as compared to two‐dimensional (2D) culture, in terms of proliferation and hsRL production, with respect to LVhsRL‐transduced cells. When implanted subcutaneously in Rankl−/− mice, these cell constructs were well tolerated, colonized by host cells, and intensely vascularized. Of note, in the bone of Rankl−/− mice that carried scaffolds with either WT or LVhsRL‐transduced Rankl−/− MSCs, we specifically observed formation of TRAP+ cells, likely due to sRL released from the scaffolds into circulation. Thus, our strategy proved to have the potential to elicit an effect on the bone; further work is required to maximize these benefits and achieve improvements of the skeletal pathology in the treated Rankl−/− mice. Stem Cells Translational Medicine2019;8:22–34
Collapse
Affiliation(s)
- Ciro Menale
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | | | - Eleonora Palagano
- Humanitas Clinical and Research Institute, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Stefano Mantero
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Marco Erreni
- Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Antonio Inforzato
- Humanitas Clinical and Research Institute, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Fontana
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Francesca Schena
- Clinica Pediatrica e Reumatologia, UOSD Centro Malattie Autoinfiammatorie e Immunodeficienze, Genoa, Italy
| | - Rob Van't Hof
- Bone Research Group, Institute of Ageing & Chronic Disease, University of Liverpool, Liverpool, UK
| | | | | | - Anna Villa
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Cristina Sobacchi
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW The term osteopetrosis refers to a group of rare skeletal diseases sharing the hallmark of a generalized increase in bone density owing to a defect in bone resorption. Osteopetrosis is clinically and genetically heterogeneous, and a precise molecular classification is relevant for prognosis and treatment. Here, we review recent data on the pathogenesis of this disorder. RECENT FINDINGS Novel mutations in known genes as well as defects in new genes have been recently reported, further expanding the spectrum of molecular defects leading to osteopetrosis. Exploitation of next-generation sequencing tools is ever spreading, facilitating differential diagnosis. Some complex phenotypes in which osteopetrosis is accompanied by additional clinical features have received a molecular classification, also involving new genes. Moreover, novel types of mutations have been recognized, which for their nature or genomic location are at high risk being neglected. Yet, the causative mutation is unknown in some patients, indicating that the genetics of osteopetrosis still deserves intense research efforts.
Collapse
Affiliation(s)
- Eleonora Palagano
- Humanitas Clinical and Research Institute, via Manzoni 113, 20089, Rozzano, MI, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Ciro Menale
- Humanitas Clinical and Research Institute, via Manzoni 113, 20089, Rozzano, MI, Italy
- Milan Unit, CNR-IRGB, Milan, Italy
| | - Cristina Sobacchi
- Humanitas Clinical and Research Institute, via Manzoni 113, 20089, Rozzano, MI, Italy.
- Milan Unit, CNR-IRGB, Milan, Italy.
| | - Anna Villa
- Humanitas Clinical and Research Institute, via Manzoni 113, 20089, Rozzano, MI, Italy
- Milan Unit, CNR-IRGB, Milan, Italy
| |
Collapse
|
21
|
Sobacchi C, Palagano E, Villa A, Menale C. Soluble Factors on Stage to Direct Mesenchymal Stem Cells Fate. Front Bioeng Biotechnol 2017; 5:32. [PMID: 28567372 PMCID: PMC5434159 DOI: 10.3389/fbioe.2017.00032] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that are identified by in vitro plastic adherence, colony-forming capacity, expression of a panel of surface molecules, and ability to differentiate at least toward osteogenic, adipogenic, and chondrogenic lineages. They also produce trophic factors with immunomodulatory, proangiogenic, and antiapoptotic functions influencing the behavior of neighboring cells. On the other hand, a reciprocal regulation takes place; in fact, MSCs can be isolated from several tissues, and depending on the original microenvironment and the range of stimuli received from there, they can display differences in their essential characteristics. Here, we focus mainly on the bone tissue and how soluble factors, such as growth factors, cytokines, and hormones, present in this microenvironment can orchestrate bone marrow-derived MSCs fate. We also briefly describe the alteration of MSCs behavior in pathological settings such as hematological cancer, bone metastasis, and bone marrow failure syndromes. Overall, the possibility to modulate MSCs plasticity makes them an attractive tool for diverse applications of tissue regeneration in cell therapy. Therefore, the comprehensive understanding of the microenvironment characteristics and components better suited to obtain a specific MSCs response can be extremely useful for clinical use.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Milan Unit, Milan, Italy.,Human Genome Laboratory, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy
| | - Eleonora Palagano
- Human Genome Laboratory, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Anna Villa
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Milan Unit, Milan, Italy.,Human Genome Laboratory, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy
| | - Ciro Menale
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Milan Unit, Milan, Italy.,Human Genome Laboratory, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy
| |
Collapse
|