1
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
2
|
Foster BL. The role of bone sialoprotein in bone healing. J Struct Biol 2024; 216:108132. [PMID: 39369971 PMCID: PMC11645215 DOI: 10.1016/j.jsb.2024.108132] [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] [Indexed: 10/08/2024]
Abstract
Bone sialoprotein (BSP) is a multi-functional extracellular matrix (ECM) protein associated with mineralized tissues, particularly bone and cementum. The amino acid sequence of BSP includes three evolutionarily conserved sequences which contribute to functions of the protein: an N-terminal collagen-binding domain, polyglutamic acid (polyE) sequences involved in hydroxyapatite nucleation and crystal growth, and a C-terminal arginine-glycine-aspartic acid (RGD) integrin-binding domain. BSP promotes attachment and differentiation of osteogenic and osteoclastic cells. Genetic ablation of BSP in mice results in skeletal and dental developmental defects and impaired bone healing in both appendicular bone and alveolar bone of the jaw. Several studies demonstrated positive effects of BSP on bone healing in rodent models, though other experiments show negligible results. Native (harvested from rat bones) BSP cross-linked to collagen induced slight improvements in calvarial bone healing in rats. Recombinant BSP and collagen delivered in a polylactide (PLA) cylinder improved bone defect healing in rat femurs. Both native and recombinant BSP delivered in a collagen gel improved alveolar bone healing in wild-type and BSP-deficient mice. These advances suggest BSP is a new player in bone healing that has potential to be an alternative or complimentary to other bioactive factors. Future studies are necessary to understand mechanisms of how BSP influences bone healing and optimize delivery and dose in different types of bone defects and injuries.
Collapse
Affiliation(s)
- B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
3
|
Kottmann V, Kolpeja E, Baumkötter G, Clauder F, Bokel A, Armbruster FP, Drees P, Gercek E, Ritz U. Bone sialoprotein stimulates cancer cell adhesion through the RGD motif and the αvβ3 and αvβ5 integrin receptors. Oncol Lett 2024; 28:542. [PMID: 39310027 PMCID: PMC11413474 DOI: 10.3892/ol.2024.14675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/02/2024] [Indexed: 09/25/2024] Open
Abstract
Being implicated in bone metastasis development, bone sialoprotein (BSP) expression is upregulated in patients with cancer. While BSP regulates cancer cell adhesion to the extracellular matrix, to the best of our knowledge, the specific adhesive molecular interactions in metastatic bone disease remain unclear. The present study aimed to improve the understanding of the arginine-glycine-aspartic acid (RGD) sequence of BSP and the integrin receptors αvβ3 and αvβ5 in BSP-mediated cancer cell adhesion. Human breast cancer (MDA-MB-231), prostate cancer (PC-3) and non-small cell lung cancer (NSCLC; NCI-H460) cell lines were cultured on BSP-coated plates. Adhesion assays with varying BSP concentrations were performed to evaluate the effect of exogenous glycine-arginine-glycine-aspartic acid-serine-proline (GRGDSP) peptide and anti-integrin antibodies on the attachment of cancer cells to BSP. Cell attachment was assessed using the alamarBlue® assay. The present results indicated that BSP supported the adhesion of cancer cells. The RGD counterpart GRGDSP peptide reduced the attachment of all tested cancer cell lines to BSP by ≤98.4%. Experiments with anti-integrin antibodies demonstrated differences among integrin receptors and cancer cell types. The αvβ5 antibody decreased NSCLC cell adhesion to BSP by 84.3%, while the αvβ3 antibody decreased adhesion by 14%. The αvβ3 antibody decreased PC-3 cell adhesion to BSP by 46.4%, while the αvβ5 antibody decreased adhesion by 9.5%. Adhesion of MDA-MB-231 cells to BSP was inhibited by 54.7% with αvβ5 antibody. The present results demonstrated that BSP-induced cancer cell adhesion occurs through the binding of the RGD sequence of BSP to the cell integrin receptors αvβ3 and αvβ5. Differences between cancer types were found regarding the mediation via αvβ3 or αvβ5 receptors. The present findings may explain why certain cancer cells preferentially spread to the bone tissue, suggesting that targeting the RGD-integrin binding interaction could be a promising novel cancer treatment option.
Collapse
Affiliation(s)
- Valentina Kottmann
- Department of Orthopaedics and Traumatology, University Medical Center of The Johannes Gutenberg University Mainz, D-55131 Mainz, Germany
| | - Elena Kolpeja
- Department of Orthopaedics and Traumatology, University Medical Center of The Johannes Gutenberg University Mainz, D-55131 Mainz, Germany
| | - Greta Baumkötter
- Department of Orthopaedics and Traumatology, University Medical Center of The Johannes Gutenberg University Mainz, D-55131 Mainz, Germany
| | | | | | | | - Philipp Drees
- Department of Orthopaedics and Traumatology, University Medical Center of The Johannes Gutenberg University Mainz, D-55131 Mainz, Germany
| | - Erol Gercek
- Department of Orthopaedics and Traumatology, University Medical Center of The Johannes Gutenberg University Mainz, D-55131 Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, University Medical Center of The Johannes Gutenberg University Mainz, D-55131 Mainz, Germany
| |
Collapse
|
4
|
Sapoznikov L, Humphrey M. Progress in Dentin-Derived Bone Graft Materials: A New Xenogeneic Dentin-Derived Material with Retained Organic Component Allows for Broader and Easier Application. Cells 2024; 13:1806. [PMID: 39513913 PMCID: PMC11544873 DOI: 10.3390/cells13211806] [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: 09/10/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
The optimal repair of rigid mineralized tissues, such as bone, in cases of fracture, surgical resection, or prosthetic placement, is a complex process often necessitating the use of bone graft materials. Autogenous bone from the patient is generally the gold standard in terms of outcomes but also has disadvantages, which have resulted in extensive research in the field of tissue engineering to develop better and more convenient alternatives. In the dental field, several initiatives have demonstrated that the dentin material derived from extracted teeth produces excellent results in terms of repairing bone defects and supporting dental implants. Dentin is acellular and thus, in contrast to autogenous bone, cannot provide osteoblasts or other cellular elements to the grafted region, but it does contain growth and differentiation factors, and has other properties that make it an impressive material for bone repair. In this review, the beneficial properties of dentin and the ways it interacts with the host bone are described in the context of bone graft materials. Autogenous tooth material has limitations, particularly in terms of the need for tooth extraction and the limited amount available, which currently restrict its use to particular dental procedures. The development of a xenograft dentin-derived material, which retains the properties of autogenous dentin, is described. Such a material could potentially enable the use of dentin-derived material more widely, particularly in orthopedic indications where its properties may be advantageous.
Collapse
|
5
|
Afsar B, Afsar RE, Caliskan Y, Lentine KL. The Relationship between Sclerostin and Kidney Transplantation Mineral Bone Disorders: A Molecule of Controversies. Calcif Tissue Int 2024; 115:339-361. [PMID: 39078512 PMCID: PMC11405501 DOI: 10.1007/s00223-024-01261-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/06/2024] [Indexed: 07/31/2024]
Abstract
Kidney transplantation is the most effective treatment option for most patients with end-stage kidney disease due to reduced mortality, decreased cardiovascular events and increased quality of life compared to patients treated with dialysis. However, kidney transplantation is not devoid of both acute and chronic complications including mineral bone disorders (MBD) which are already present in patients with chronic kidney disease (CKD) before kidney transplantation. The natural history of MBD after kidney transplantation is variable and new markers are needed to define MBD after kidney transplantation. One of these promising molecules is sclerostin. The main action of sclerostin is to inhibit bone formation and mineralization by blocking osteoblast differentiation and function. In kidney transplant recipients (KTRs), various studies have shown that sclerostin is associated with graft function, bone parameters, vascular calcification, and arterial stiffness although non-uniformly. Furthermore, data for inhibition of sclerostin with monoclonal antibody romosozumab for treatment of osteoporosis is available for general population but not in KTRs which osteoporosis is highly prevalent. In this narrative review, we have summarized the studies investigating the change of sclerostin before and after kidney transplantation, the relationship between sclerostin and laboratory parameters, bone metabolism and vascular calcification in the context of kidney transplantation. We also pointed out the uncertainties, explained the causes of divergent findings and suggest further potential study topics regarding sclerostin in kidney transplantation.
Collapse
Affiliation(s)
- Baris Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey.
- Department of Nephrology, Saint Loui University, Saint Louis University Hospital, Saint Louis, MO, USA.
| | - Rengin Elsurer Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey
- Department of Nephrology, Saint Loui University, Saint Louis University Hospital, Saint Louis, MO, USA
| | - Yasar Caliskan
- Department of Nephrology, Saint Loui University, Saint Louis University Hospital, Saint Louis, MO, USA
| | - Krista L Lentine
- Department of Nephrology, Saint Loui University, Saint Louis University Hospital, Saint Louis, MO, USA
| |
Collapse
|
6
|
Ganapathy A, Narayanan K, Chen Y, Villani C, George A. Dentin matrix protein 1 and HUVEC-ECM scaffold promote the differentiation of human dental pulp stem cells into endothelial lineage: implications in regenerative medicine. Front Physiol 2024; 15:1429247. [PMID: 39040080 PMCID: PMC11260688 DOI: 10.3389/fphys.2024.1429247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Reprograming of the dental pulp somatic cells to endothelial cells is an attractive strategy for generation of new blood vessels. For tissue regeneration, vascularization of engineered constructs is crucial to improve repair mechanisms. In this study, we show that dentin matrix protein 1 (DMP1) and HUVEC-ECM scaffold enhances the differentiation potential of dental pulp stem cells (DPSCs) to an endothelial phenotype. Our results show that the differentiated DPSCs expressed endothelial markers CD31 and VE-Cadherin (CD144) at 7 and 14 days. Expression of CD31 and VE-Cadherin (CD144) were also confirmed by immunofluorescence. Furthermore, flow cytometry analysis revealed a steady increase in CD31 and VE-Cadherin (CD144) positive cells with DMP1 treatment when compared with control. In addition, integrins specific for endothelial cells were highly expressed during the differentiation process. The endothelial cell signature of differentiated DPSCs were additionally characterized for key endothelial cell markers using gene expression by RT-PCR, Western blotting, immunostaining, and RNA-seq analysis. Furthermore, the angiogenic phenotype was confirmed by tubule and capillary sprout formation. Overall, stimulation of DPSCs by DMP1 and use of HUVEC-ECM scaffold promoted their differentiation into phenotypically, transcriptionally, and functionally differentiated bonafide endothelial cells. This study is novel, physiologically relevant and different from conventional strategies.
Collapse
Affiliation(s)
| | | | | | | | - Anne George
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
7
|
Mao M, Ahrens L, Luka J, Contreras F, Kurkina T, Bienstein M, Sárria Pereira de Passos M, Schirinzi G, Mehn D, Valsesia A, Desmet C, Serra MÁ, Gilliland D, Schwaneberg U. Material-specific binding peptides empower sustainable innovations in plant health, biocatalysis, medicine and microplastic quantification. Chem Soc Rev 2024; 53:6445-6510. [PMID: 38747901 DOI: 10.1039/d2cs00991a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Material-binding peptides (MBPs) have emerged as a diverse and innovation-enabling class of peptides in applications such as plant-/human health, immobilization of catalysts, bioactive coatings, accelerated polymer degradation and analytics for micro-/nanoplastics quantification. Progress has been fuelled by recent advancements in protein engineering methodologies and advances in computational and analytical methodologies, which allow the design of, for instance, material-specific MBPs with fine-tuned binding strength for numerous demands in material science applications. A genetic or chemical conjugation of second (biological, chemical or physical property-changing) functionality to MBPs empowers the design of advanced (hybrid) materials, bioactive coatings and analytical tools. In this review, we provide a comprehensive overview comprising naturally occurring MBPs and their function in nature, binding properties of short man-made MBPs (<20 amino acids) mainly obtained from phage-display libraries, and medium-sized binding peptides (20-100 amino acids) that have been reported to bind to metals, polymers or other industrially produced materials. The goal of this review is to provide an in-depth understanding of molecular interactions between materials and material-specific binding peptides, and thereby empower the use of MBPs in material science applications. Protein engineering methodologies and selected examples to tailor MBPs toward applications in agriculture with a focus on plant health, biocatalysis, medicine and environmental monitoring serve as examples of the transformative power of MBPs for various industrial applications. An emphasis will be given to MBPs' role in detecting and quantifying microplastics in high throughput, distinguishing microplastics from other environmental particles, and thereby assisting to close an analytical gap in food safety and monitoring of environmental plastic pollution. In essence, this review aims to provide an overview among researchers from diverse disciplines in respect to material-(specific) binding of MBPs, protein engineering methodologies to tailor their properties to application demands, re-engineering for material science applications using MBPs, and thereby inspire researchers to employ MBPs in their research.
Collapse
Affiliation(s)
- Maochao Mao
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Leon Ahrens
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Julian Luka
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Francisca Contreras
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Tetiana Kurkina
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Marian Bienstein
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | | | | | - Dora Mehn
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Andrea Valsesia
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Cloé Desmet
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | | | - Ulrich Schwaneberg
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| |
Collapse
|
8
|
Islam P, Ice JA, Alake SE, Adedigba P, Hatter B, Robinson K, Clarke SL, Ford Versypt AN, Ritchey J, Lucas EA, Smith BJ. Fructooligosaccharides act on the gut-bone axis to improve bone independent of Tregs and alter osteocytes in young adult C57BL/6 female mice. JBMR Plus 2024; 8:ziae021. [PMID: 38562914 PMCID: PMC10982850 DOI: 10.1093/jbmrpl/ziae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/15/2023] [Accepted: 01/20/2024] [Indexed: 04/04/2024] Open
Abstract
Targeting the gut-bone axis with probiotics and prebiotics is considered as a promising strategy to reduce the risk of osteoporosis. Gut-derived short chain fatty acids (SCFA) mediate the effects of probiotics on bone via Tregs, but it is not known whether prebiotics act through a similar mechanism. We investigated how 2 different prebiotics, tart cherry (TC) and fructooligosaccharide (FOS), affect bone, and whether Tregs are required for this response. Eight-wk-old C57BL/6 female mice were fed with diets supplemented with 10% w/w TC, FOS, or a control diet (Con; AIN-93M) diet, and they received an isotype control or CD25 Ab to suppress Tregs. The FOS diet increased BMC, density, and trabecular bone volume in the vertebra (~40%) and proximal tibia (~30%) compared to the TC and control diets (Con), irrespective of CD25 treatment. Both prebiotics increased (P < .01) fecal SCFAs, but the response was greater with FOS. To determine how FOS affected bone cells, we examined genes involved in osteoblast and osteoclast differentiation and activity as well as genes expressed by osteocytes. The FOS increased the expression of regulators of osteoblast differentiation (bone morphogenetic protein 2 [Bmp2], Wnt family member 10b [Wnt10b] and Osterix [Osx]) and type 1 collagen). Osteoclasts regulators were unaltered. The FOS also increased the expression of genes associated with osteocytes, including (Phex), matrix extracellular phosphoglycoprotein (Mepe), and dentin matrix acidic phosphoprotein 1 (Dmp-1). However, Sost, the gene that encodes for sclerostin was also increased by FOS as the number and density of osteocytes increased. These findings demonstrate that FOS has a greater effect on the bone mass and structure in young adult female mice than TC and that its influence on osteoblasts and osteocytes is not dependent on Tregs.
Collapse
Affiliation(s)
- Proapa Islam
- Nutritional Sciences Department, Oklahoma State University, Stillwater, OK 74078, USA
| | - John A Ice
- Nutritional Sciences Department, Oklahoma State University, Stillwater, OK 74078, USA
| | - Sanmi E Alake
- Nutritional Sciences Department, Oklahoma State University, Stillwater, OK 74078, USA
| | - Pelumi Adedigba
- Indiana Center for Musculoskeletal Health, Indiana School of Medicine, Indianapolis, IN 46202, USA
| | - Bethany Hatter
- Nutritional Sciences Department, Oklahoma State University, Stillwater, OK 74078, USA
| | - Kara Robinson
- Nutritional Sciences Department, Oklahoma State University, Stillwater, OK 74078, USA
| | - Stephen L Clarke
- Nutritional Sciences Department, Oklahoma State University, Stillwater, OK 74078, USA
| | - Ashlee N Ford Versypt
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260, USA
| | - Jerry Ritchey
- Veterinary Pathobiology Department, Oklahoma State University, Stillwater, OK 74078, USA
| | - Edralin A Lucas
- Nutritional Sciences Department, Oklahoma State University, Stillwater, OK 74078, USA
| | - Brenda J Smith
- Indiana Center for Musculoskeletal Health, Indiana School of Medicine, Indianapolis, IN 46202, USA
- Department of Obstetrics and Gynecology, Indiana School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
9
|
Nguyen NTK, Lee SS, Chen PH, Chang YH, Pham NN, Chang CW, Pham DH, Ngo DKT, Dang QT, Truong VA, Truong VA, Chang YH, Hu YC. Enhanced Calvarial Bone Repair Using ASCs Engineered with RNA-Guided Split dCas12a System that Co-Activates Sox 5, Sox6, and Long Non-Coding RNA H19. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306612. [PMID: 38126683 DOI: 10.1002/smll.202306612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/16/2023] [Indexed: 12/23/2023]
Abstract
Healing of large calvarial bone defects remains challenging. An RNA-guided Split dCas12a system is previously harnessed to activate long non-coding RNA H19 (lncRNA H19, referred to as H19 thereafter) in bone marrow-derived mesenchymal stem cells (BMSCs). H19 activation in BMSCs induces chondrogenic differentiation, switches bone healing pathways, and improves calvarial bone repair. Since adipose-derived stem cells (ASCs) can be harvested more easily in large quantity, here it is aimed to use ASCs as an alternative cell source. However, H19 activation alone using the Split dCas12a system in ASCs failed to elicit evident chondrogenesis. Therefore, split dCas12a activators are designed more to co-activate other chondroinductive transcription factors (Sox5, Sox6, and Sox9) to synergistically potentiate differentiation. It is found that co-activation of H19/Sox5/Sox6 in ASCs elicited more potent chondrogenic differentiation than activation of Sox5/Sox6/Sox9 or H19 alone. Co-activating H19/Sox5/Sox6 in ASCs significantly augmented in vitro cartilage formation and in vivo calvarial bone healing. These data altogether implicated the potentials of the Split dCas12a system to trigger multiplexed gene activation in ASCs for differentiation pathway reprogramming and tissue regeneration.
Collapse
Affiliation(s)
- Nuong Thi Kieu Nguyen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Shang-Shung Lee
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Pin-Hsin Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Yi-Hao Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Nam Ngoc Pham
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Chin-Wei Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Dang Huu Pham
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Dung Kim Thi Ngo
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Quyen Thuc Dang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Vy Anh Truong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Vu Anh Truong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Yu-Han Chang
- Department of Orthopaedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, 33305, Taiwan
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 300044, Taiwan
| |
Collapse
|
10
|
Sawada K, Shimomura J, Takedachi M, Murata M, Morimoto C, Kawasaki K, Kawakami K, Iwayama T, Murakami S. Activation of periodontal ligament cell cytodifferentiation by juxtacrine signaling from cementoblasts. J Periodontol 2024; 95:256-267. [PMID: 37492992 DOI: 10.1002/jper.23-0211] [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: 03/25/2023] [Revised: 06/12/2023] [Accepted: 07/22/2023] [Indexed: 07/27/2023]
Abstract
BACKGROUND New cementum forms from existing cementum during periodontal tissue regeneration, indicating that cementoblasts may interact with progenitor cells in the periodontal ligament to enhance cementogenesis. However, the molecular mechanisms of this process are currently unknown. This study aims to clarify the role of cell-cell interactions between cementoblasts and periodontal ligament cells in differentiation into cementoblasts. METHODS To analyze the role of human cementoblast-like cells (HCEMs) on human periodontal ligament cells (HPDLs), we mixed cell suspensions of enhanced green fluorescent protein-tagged HPDLs and HCEMs, and then seeded and cultured them in single wells (direct co-cultures). We sorted co-cultured HPDLs and analyzed their characteristics, including the expression of cementum-related genes. In addition, we cultured HPDLs and HCEMs in a non-contact environment using a culture system composed of an upper insert and a lower well separated by a semi-permeable membrane (indirect co-cultures), and similar analysis was performed. Gene expression of integrin-binding sialoprotein (IBSP) in cementoblasts was confirmed in mouse periodontal tissues. We also investigated the effect of Wingless-type (Wnt) signaling on the differentiation of HPDLs into cementoblasts. RESULTS Direct co-culture of HPDLs with HCEMs significantly upregulated the expression of cementoblast-related genes in HPDLs, whereas indirect co-culture exerted no effect. Wnt3A stimulation significantly upregulated IBSP expression in HPDLs, whereas inhibition of canonical Wnt signaling suppressed the effects of co-culture. CONCLUSION Our results suggest that direct cell interactions with cementoblasts promote periodontal ligament cell differentiation into cementoblasts. Juxtacrine signaling via the canonical Wnt pathway plays a role in this interaction.
Collapse
Affiliation(s)
- Keigo Sawada
- Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Junpei Shimomura
- Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Masahide Takedachi
- Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Mari Murata
- Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Chiaki Morimoto
- Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Kohsuke Kawasaki
- Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Kazuma Kawakami
- Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Tomoaki Iwayama
- Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Shinya Murakami
- Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| |
Collapse
|
11
|
Kaur S, Roberts DD. Why do humans need thrombospondin-1? J Cell Commun Signal 2023; 17:485-493. [PMID: 36689135 PMCID: PMC10409698 DOI: 10.1007/s12079-023-00722-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/07/2023] [Indexed: 01/24/2023] Open
Abstract
Matricellular proteins comprise several families of secreted proteins that function in higher animals at the interface between cells and their surrounding extracellular matrix. Targeted gene disruptions that result in loss of viability in mice have revealed critical roles for several matricellular proteins in murine embryonic development, including two members of the cellular communication network (CCN) gene family. In contrast, mice lacking single or multiple members of the thrombospondin (THBS) gene family remain viable and fertile. The frequency of loss of function mutants, identified using human deep exome sequencing data, provided evidence that some of the essential genes in mice, including Ccn1, are also essential genes in humans. However, a deficit in loss of function mutants in humans indicated that THBS1 is also highly loss-intolerant. In addition to roles in embryonic development or adult reproduction, genes may be loss-intolerant in humans because their function is needed to survive environmental stresses that are encountered between birth and reproduction. Laboratory mice live in a protected environment that lacks the exposures to pathogens and injury that humans routinely face. However, subjecting Thbs1-/- mice to defined stresses has provided valuable insights into functions of thrombospondin-1 that could account for the loss-intolerance of THBS1 in humans. Stress response models using transgenic mice have identified protective functions of thrombospondin-1 in the cardiovascular system (red) and immune defenses (blue) that could account for its intolerance to loss of function mutants in humans.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 Room 2S235, 10 Center Dr, Bethesda, MD, 20892-1500, USA
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 Room 2S235, 10 Center Dr, Bethesda, MD, 20892-1500, USA.
| |
Collapse
|
12
|
Amorim S, Dudik O, Soares da Costa D, Reis RL, Silva TH, Pires RA. Fucoidan-Coated Silica Nanoparticles Promote the Differentiation of Human Mesenchymal Stem Cells into the Osteogenic Lineage. ACS Biomater Sci Eng 2023; 9:4907-4915. [PMID: 37493090 DOI: 10.1021/acsbiomaterials.3c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Silica nanoparticles (SiNPs) are widely used in biomedical applications, such as cancer therapy/diagnosis or tissue engineering and regenerative medicine. Herein, we synthesized SiNPs and modified them with sulfonic acid groups (by organosilylation followed by oxidation) or a sulfated polysaccharide (i.e., fucoidan, a seaweed biopolymer, by using electrostatic surface immobilization) due to the known capacity of the sulfonic/sulfate moieties to stabilize proteins and promote stem cell differentiation toward the osteogenic lineage. The developed pristine and functionalized nanoparticles were characterized by dynamic light scattering (DLS), scanning electron microscopy (SEM), transmission electron microscopy (TEM), and X-ray photoelectron spectroscopy (XPS), showing the monodisperse size distribution (between 360 and 450 nm) and the success of the coating/functionalization with fucoidan or sulfonic groups. The developed SiNPs (at a concentration of 50 μg/mL) were assessed through their contact with SaOs2 cells evidencing their cytocompatibility. Furthermore, the osteogenic differentiation of bmMSCs was evaluated by the quantification of ALP activity, as well as the expression profile of osteogenic-related genes, such as Runx2, ALP, and OP. We found that the coating of the SiNPs with fucoidan induced the osteogenic differentiation of bmMSCs, being an effective mediator of bone regeneration.
Collapse
Affiliation(s)
- Sara Amorim
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| | - Olesia Dudik
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| | - Diana Soares da Costa
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| | - Tiago H Silva
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| | - Ricardo A Pires
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| |
Collapse
|
13
|
Mae T, Hasegawa T, Hongo H, Yamamoto T, Zhao S, Li M, Yamazaki Y, Amizuka N. Immunolocalization of Enzymes/Membrane Transporters Related to Bone Mineralization in the Metaphyses of the Long Bones of Parathyroid-Hormone-Administered Mice. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1179. [PMID: 37374382 DOI: 10.3390/medicina59061179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
The present study aimed to demonstrate the immunolocalization and/or gene expressions of the enzymes and membrane transporters involved in bone mineralization after the intermittent administration of parathyroid hormone (PTH). The study especially focused on TNALP, ENPP1, and PHOSPHO1, which are involved in matrix vesicle-mediated mineralization, as well as PHEX and the SIBLING family, which regulate mineralization deep inside bone. Six-week-old male mice were subcutaneously injected with 20 μg/kg/day of human PTH (1-34) two times per day (n = 6) or four times per day (n = 6) for two weeks. Additionally, control mice (n = 6) received a vehicle. Consistently with an increase in the volume of the femoral trabeculae, the mineral appositional rate increased after PTH administration. The areas positive for PHOSPHO1, TNALP, and ENPP1 in the femoral metaphyses expanded, and the gene expressions assessed by real-time PCR were elevated in PTH-administered specimens when compared with the findings in control specimens. The immunoreactivity and/or gene expressions of PHEX and the SIBLING family (MEPE, osteopontin, and DMP1) significantly increased after PTH administration. For example, MEPE immunoreactivity was evident in some osteocytes in PTH-administered specimens but was hardly observed in control specimens. In contrast, mRNA encoding cathepsin B was significantly reduced. Therefore, the bone matrix deep inside might be further mineralized by PHEX/SIBLING family after PTH administration. In summary, it is likely that PTH accelerates mineralization to maintain a balance with elevated matrix synthesis, presumably by mediating TNALP/ENPP1 cooperation and stimulating PHEX/SIBLING family expression.
Collapse
Affiliation(s)
- Takahito Mae
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
- Department of Gerontology, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| | - Tomoka Hasegawa
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| | - Hiromi Hongo
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| | - Tomomaya Yamamoto
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
- Northern Army Medical Unit, Camp Makomanai, Japan Ground Self-Defense Forces, Sapporo 005-8543, Japan
| | - Shen Zhao
- Department of Endodontics and Operative Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Minqi Li
- Center of Osteoporosis and Bone Mineral Research, Department of Bone Metabolism, School of Stomatology, Shandong University, Jinan 250012, China
| | - Yutaka Yamazaki
- Department of Gerontology, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| | - Norio Amizuka
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| |
Collapse
|
14
|
Palma-Lara I, García Alonso-Themann P, Pérez-Durán J, Godínez-Aguilar R, Bonilla-Delgado J, Gómez-Archila D, Espinosa-García AM, Nolasco-Quiroga M, Victoria-Acosta G, López-Ornelas A, Serrano-Bello JC, Olguín-García MG, Palacios-Reyes C. Potential Role of Protein Kinase FAM20C on the Brain in Raine Syndrome, an In Silico Analysis. Int J Mol Sci 2023; 24:ijms24108904. [PMID: 37240249 DOI: 10.3390/ijms24108904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
FAM20C (family with sequence similarity 20, member C) is a serine/threonine-specific protein kinase that is ubiquitously expressed and mainly associated with biomineralization and phosphatemia regulation. It is mostly known due to pathogenic variants causing its deficiency, which results in Raine syndrome (RNS), a sclerosing bone dysplasia with hypophosphatemia. The phenotype is recognized by the skeletal features, which are related to hypophosphorylation of different FAM20C bone-target proteins. However, FAM20C has many targets, including brain proteins and the cerebrospinal fluid phosphoproteome. Individuals with RNS can have developmental delay, intellectual disability, seizures, and structural brain defects, but little is known about FAM20C brain-target-protein dysregulation or about a potential pathogenesis associated with neurologic features. In order to identify the potential FAM20C actions on the brain, an in silico analysis was conducted. Structural and functional defects reported in RNS were described; FAM20C targets and interactors were identified, including their brain expression. Gene ontology of molecular processes, function, and components was completed for these targets, as well as for potential involved signaling pathways and diseases. The BioGRID and Human Protein Atlas databases, the Gorilla tool, and the PANTHER and DisGeNET databases were used. Results show that genes with high expression in the brain are involved in cholesterol and lipoprotein processes, plus axo-dendritic transport and the neuron part. These results could highlight some proteins involved in the neurologic pathogenesis of RNS.
Collapse
Affiliation(s)
- Icela Palma-Lara
- Laboratorio de Morfología Celular y Molecular, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | | | - Javier Pérez-Durán
- Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México 11000, Mexico
| | | | - José Bonilla-Delgado
- Unidad de Investigación, Hospital Regional de Ixtapaluca, Ixtapaluca 56530, Mexico
- Departamento de Biotecnología, Escuela de Ingeniería y Ciencias, Instituto Tecnológico de Monterrey, Toluca de Lerdo 50110, Mexico
| | - Damián Gómez-Archila
- Departamento de Oncología Quirúrgica, Hospital de Gineco-Obstetricia 3, Centro Médico Nacional "La Raza", Ciudad de México 02990, Mexico
| | | | - Manuel Nolasco-Quiroga
- Coordinación de Enseñanza e Investigación, Clínica Hospital Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Huauchinango 73177, Mexico
| | | | - Adolfo López-Ornelas
- División de Investigación, Hospital Juárez de México, Ciudad de México 11340, Mexico
| | - Juan Carlos Serrano-Bello
- Departamento de Patología Clínica y Experimental, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico
| | | | - Carmen Palacios-Reyes
- División de Investigación, Hospital Juárez de México, Ciudad de México 11340, Mexico
| |
Collapse
|
15
|
Elias J, Matheson BA, Gower L. Influence of Crosslinking Methods on Biomimetically Mineralized Collagen Matrices for Bone-like Biomaterials. Polymers (Basel) 2023; 15:polym15091981. [PMID: 37177129 PMCID: PMC10180878 DOI: 10.3390/polym15091981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
To assist in bone defect repair, ideal bone regeneration scaffolds should exhibit good osteoconductivity and osteoinductivity, but for load-bearing applications, they should also have mechanical properties that emulate those of native bone. The use of biomimetic processing methods for the mineralization of collagen fibrils has resulted in interpenetrating composites that mimic the nanostructure of native bone; however, closely matching the mechanical properties of bone on a larger scale is something that is still yet to be achieved. In this study, four different collagen crosslinking methods (EDC-NHS, quercetin, methacrylated collagen, and riboflavin) are compared and combined with biomimetic mineralization via the polymer-induced liquid-precursor (PILP) process, to obtain bone-like collagen-hydroxyapatite composites. Densified fibrillar collagen scaffolds were fabricated, crosslinked, and biomimetically mineralized using the PILP process, and the effect of each crosslinking method on the degree of mineralization, tensile strength, and modulus of the mineralized scaffolds were analyzed and compared. Improved modulus and tensile strength values were obtained using EDC-NHS and riboflavin crosslinking methods, while quercetin and methacrylated collagen resulted in little to no increase in mechanical properties. Decreased mineral contents appear to be necessary for retaining tensile strength, suggesting that mineral content should be kept below a percolation threshold to optimize properties of these interpenetrating nanocomposites. This work supports the premise that a combination of collagen crosslinking and biomimetic mineralization methods may provide solutions for fabricating robust bone-like composites on a larger scale.
Collapse
Affiliation(s)
- Jeremy Elias
- Department of Materials Science & Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Bobbi-Ann Matheson
- Department of Materials Science & Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Laurie Gower
- Department of Materials Science & Engineering, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
16
|
Markovics A, Lupo S, Patel N, Mikecz K, Sumner DR, Ross RD. SHP-1 Protein Tyrosine Phosphatase Affects Early Postnatal Bone Development in Mice. Calcif Tissue Int 2023; 112:472-482. [PMID: 36725700 DOI: 10.1007/s00223-023-01064-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/20/2023] [Indexed: 02/03/2023]
Abstract
The Src homology region 2 domain-containing phosphatase-1 (SHP-1) is an intracellular tyrosine phosphatase that plays a negative regulatory role in immune cell signaling. Absent or diminished SHP-1 catalytic activity results in reduced bone mass with enhanced bone resorption. Here, we sought to investigate if Shp1 overexpression leads to increased bone mass and improved mechanical properties. Male and female wildtype (WT) and SHP1-transgenic (Tg) mice at 28, 56, and 84 days of age were compared. We applied microcomputed tomography to assess femoral cortical bone geometry and trabecular architecture and 3-point mechanical bending to assess mid-diaphyseal structural and estimated material properties. Serum OPG, RANKL, P1NP, and CTX-1 concentrations were measured by enzyme-linked immunoassay. The majority of transgene effects were restricted to the 28-day-old mice. Trabecular bone volume per total volume, trabecular number, and connectivity density were greater in 28-day-old female SHP1-Tg mice when compared to WTs. SHP1-Tg female mice showed increased total and medullary areas, with no difference in cortical area and thickness. Cortical tissue mineral density was strongly genotype-dependent. Failure load, yield load, ultimate stress, and yield stress were all lower in 28-day-old SHP1-Tg females. In 28-day-old SHP1-Tg females, circulating levels of OPG and P1NP were higher and RANKL levels were lower than WT controls. Our study demonstrates a role for SHP-1 in early postnatal bone development; SHP-1 overexpression negatively impacted whole bone strength and material properties in females.
Collapse
Affiliation(s)
- Adrienn Markovics
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA.
| | - Sydney Lupo
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Niyati Patel
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Katalin Mikecz
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - D Rick Sumner
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, USA
| | - Ryan D Ross
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
17
|
Ni X, Gong Y, Jiang Y, Li X, Pang Q, Liu W, Chi Y, Jiajue R, Wang O, Li M, Xing X, Xia W. The First Compound Heterozygous Mutations of DMP1 Causing Rare Autosomal Recessive Hypophosphatemic Rickets Type 1. J Clin Endocrinol Metab 2023; 108:791-801. [PMID: 36334264 DOI: 10.1210/clinem/dgac640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/11/2022] [Indexed: 11/08/2022]
Abstract
CONTEXT Hereditary hypophosphatemic rickets (HR) consists of a group of inherited hypophosphatemia due to mutations of different genes, which need genetic analysis to make a differential diagnosis. Among them, autosomal recessive hypophosphatemic rickets type 1 (ARHR1), caused by a homozygous mutation of dentin matrix protein 1 (DMP1), is extremely rare, with only 30 reported patients. To date, there has been no case with compound heterozygous DMP1 mutations. OBJECTIVE To report the first compound heterozygous mutations of DMP1 causing ARHR1 and confirm the effect of the mutation on DMP1 protein. METHODS We report the clinical features of a Chinese patient with HR. Whole-exome sequencing (WES) was performed on the proband. Then, Cytoscan HD array, Sanger sequencing, and genomic quantitative PCR (qPCR) were used to confirm the mutations. A cell experiment was conducted to explore the effect of the mutation. RESULTS The proband is a 4-year-old boy, who developed genu varum when he was able to walk at age 1 year and tooth loss after a mild hit at age 3.5 years. Physical examination, biochemical measurement, and imaging finding indicated HR. Family history was negative. WES performed on the proband revealed a novel start codon mutation (c.1A > T, p.Met1Leu) in DMP1 and a large deletion involving most of the small integrin-binding ligand N-linked glycoprotein (SIBLING) family gene, including DSPP, DMP1, IBSP, and MEPE. The novel paternally inherited start codon mutation, which resulted in decreased expression of DMP1 protein with smaller molecular weight and cleavage defect, was confirmed by Sanger sequencing. The maternally inherited deletion was validated by Cytoscan and qPCR, and the breakpoint was finally identified by long-range PCR and Sanger sequencing. Manifestation of dentin dysplasia (DD) or dentinogenesis imperfecta (DGI) caused by DSPP mutations was absent in the patient and his mother, confirming that haploinsufficiency could not lead to DD or DGI. CONCLUSION We report for the first time compound heterozygous DMP1 mutations consisting of a large deletion and a novel start codon mutation (c.1A > T, p.Met1Leu) in a Chinese patient with ARHR1.
Collapse
Affiliation(s)
- Xiaolin Ni
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yiyi Gong
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yan Jiang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiang Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qianqian Pang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Wei Liu
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yue Chi
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ruizhi Jiajue
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ou Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Mei Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiaoping Xing
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Weibo Xia
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
18
|
Osteopontin: A Bone-Derived Protein Involved in Rheumatoid Arthritis and Osteoarthritis Immunopathology. Biomolecules 2023; 13:biom13030502. [PMID: 36979437 PMCID: PMC10046882 DOI: 10.3390/biom13030502] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Osteopontin (OPN) is a bone-derived phosphoglycoprotein related to physiological and pathological mechanisms that nowadays has gained relevance due to its role in the immune system response to chronic degenerative diseases, including rheumatoid arthritis (RA) and osteoarthritis (OA). OPN is an extracellular matrix (ECM) glycoprotein that plays a critical role in bone remodeling. Therefore, it is an effector molecule that promotes joint and cartilage destruction observed in clinical studies, in vitro assays, and animal models of RA and OA. Since OPN undergoes multiple modifications, including posttranslational changes, proteolytic cleavage, and binding to a wide range of receptors, the mechanisms by which it produces its effects, in some cases, remain unclear. Although there is strong evidence that OPN contributes significantly to the immunopathology of RA and OA when considering it as a common denominator molecule, some experimental trial results argue for its protective role in rheumatic diseases. Elucidating in detail OPN involvement in bone and cartilage degeneration is of interest to the field of rheumatology. This review aims to provide evidence of the OPN’s multifaceted role in promoting joint and cartilage destruction and propose it as a common denominator of AR and OA immunopathology.
Collapse
|
19
|
Bone Sialoprotein Immobilized in Collagen Type I Enhances Angiogenesis In Vitro and In Ovo. Polymers (Basel) 2023; 15:polym15041007. [PMID: 36850289 PMCID: PMC9968013 DOI: 10.3390/polym15041007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/24/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Bone fracture healing is a multistep process, including early immunological reactions, osteogenesis, and as a key factor, angiogenesis. Molecules inducing osteogenesis as well as angiogenesis are rare, but hold promise to be employed in bone tissue engineering. It has been demonstrated that the bone sialoprotein (BSP) can induce bone formation when immobilized in collagen type I, but its effect on angiogenesis still has to be characterized in detail. Therefore, the aim of this study was to analyse the effects of BSP immobilized in a collagen type I gel on angiogenesis. First, in vitro analyses with endothelial cells (HUVECs) were performed detecting enhancing effects of BSP on proliferation and gene expression of endothelial markers. A spheroid model was employed confirming these results. Finally, the inducing impact of BSP-collagen on vascular density was proved in a yolk sac membrane assay. Our results demonstrate that BSP is capable of inducing angiogenesis and confirm that collagen type I is the optimal carrier for this protein. Taking into account former results, and literature showing that BSP also induces osteogenesis, one can hypothesize that BSP couples angiogenesis and osteogenesis, making it a promising molecule to be used in bone tissue regeneration.
Collapse
|
20
|
Histological Assessment of Endochondral Ossification and Bone Mineralization. ENDOCRINES 2023. [DOI: 10.3390/endocrines4010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Finely tuned cartilage mineralization, endochondral ossification, and normal bone formation are necessary for normal bone growth. Hypertrophic chondrocytes in the epiphyseal cartilage secrete matrix vesicles, which are small extracellular vesicles initiating mineralization, into the intercolumnar septa but not the transverse partitions of the cartilage columns. Bone-specific blood vessels invade the unmineralized transverse septum, exposing the mineralized cartilage cores. Many osteoblast precursors migrate to the cartilage cores, where they synthesize abundant bone matrices, and mineralize them in a process of matrix vesicle-mediated bone mineralization. Matrix vesicle-mediated mineralization concentrates calcium (Ca) and inorganic phosphates (Pi), which are converted into hydroxyapatite crystals. These crystals grow radially and are eventually get out of the vesicles to form spherical mineralized nodules, leading to collagen mineralization. The influx of Ca and Pi into the matrix vesicle is regulated by several enzymes and transporters such as TNAP, ENPP1, PiT1, PHOSPHO1, annexins, and others. Such matrix vesicle-mediated mineralization is regulated by osteoblastic activities, synchronizing the synthesis of organic bone material. However, osteocytes reportedly regulate peripheral mineralization, e.g., osteocytic osteolysis. The interplay between cartilage mineralization and vascular invasion during endochondral ossification, as well as that of osteoblasts and osteocytes for normal mineralization, appears to be crucial for normal bone growth.
Collapse
|
21
|
Anada R, Hara ES, Nagaoka N, Okada M, Kamioka H, Matsumoto T. Important roles of odontoblast membrane phospholipids in early dentin mineralization. J Mater Chem B 2023; 11:657-666. [PMID: 36541228 DOI: 10.1039/d2tb02351b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The objective of this study was to first identify the timing and location of early mineralization of mouse first molar, and subsequently, to characterize the nucleation site for mineral formation in dentin from a materials science viewpoint and evaluate the effect of environmental cues (pH) affecting early dentin formation. Early dentin mineralization in mouse first molars began in the buccal central cusp on post-natal day 0 (P0), and was first hypothesized to involve collagen fibers. However, elemental mapping indicated the co-localization of phospholipids with collagen fibers in the early mineralization area. Co-localization of phosphatidylserine and annexin V, a functional protein that binds to plasma membrane phospholipids, indicated that phospholipids in the pre-dentin matrix were derived from the plasma membrane. A 3-dimensional in vitro biomimetic mineralization assay confirmed that phospholipids from the plasma membrane are critical factors initiating mineralization. Additionally, the direct measurement of the tooth germ pH, indicated it to be alkaline. The alkaline environment markedly enhanced the mineralization of cell membrane phospholipids. These results indicate that cell membrane phospholipids are nucleation sites for mineral formation, and could be important materials for bottom-up approaches aiming for rapid and more complex fabrication of dentin-like structures.
Collapse
Affiliation(s)
- Risa Anada
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan. .,Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Emilio Satoshi Hara
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| | - Noriyuki Nagaoka
- Advanced Research Center for Oral and Craniofacial Sciences, Dental School, Okayama University, Okayama, Japan
| | - Masahiro Okada
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| |
Collapse
|
22
|
Calsa B, Bortolança TJ, Masiero BC, Esquisatto MAM, de Oliveira CA, Catisti R, Santamaria-Jr M. Maxillary and dental development in the offspring of protein-restricted female rats. Eur J Oral Sci 2022; 130:e12895. [PMID: 36199171 DOI: 10.1111/eos.12895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/31/2022] [Indexed: 12/13/2022]
Abstract
Nutritional restriction during developmental periods impairs organ physiology. Female rats were subjected to protein restriction during pregnancy and lactation to analyze dental and maxillary development. Four exposure groups were considered: normal-protein diet during pregnancy and lactation (NP, 17% casein), low-protein diet during lactation (LP-L, 6% casein), low-protein diet during pregnancy and lactation (LP), and low-protein diet during pregnancy (LP-G). Maxillae from 15-day-old male pups were collected. All protein-restricted groups presented increased dentin thickness and reduced alveolar bone area. When protein restriction was applied during both gestation and lactation (LP), harmful effects were observed in the form of loss of protective OPG (osteoprotegerin) in tooth epithelium-mesenchyme, due to higher RANKL expression, delay in odontoblast maturation, less dental pulp vascularity, reduction in amount of alveolar bone, and less matrix mineralization. In the LP-L group, effects of protein restriction seemed less harmful, and despite less alveolar bone, the enhancement in BMP-7, VEGF, and RANKL seems a compensatory signal to maintain maxillary osteogenesis. In LP-G animals, Dspp expression was higher, suggesting a delay in odontoblast maturation or expression recuperation. In conclusion, maternal protein restriction affects dental and maxillary development. A low-protein diet only in gestation allows for normal development. A low-protein diet during gestation-lactation results in impaired odontogenesis that may increase susceptibility of dental anomalies.
Collapse
Affiliation(s)
- Bruno Calsa
- Graduate Program of Biomedical Sciences, Herminio Ometto University Center, Araras, São Paulo, Brazil
| | | | - Beatriz Calloni Masiero
- Graduate Program of Biomedical Sciences, Herminio Ometto University Center, Araras, São Paulo, Brazil
| | | | - Camila Andrea de Oliveira
- Graduate Program of Biomedical Sciences, Herminio Ometto University Center, Araras, São Paulo, Brazil
| | - Rosana Catisti
- Graduate Program of Biomedical Sciences, Herminio Ometto University Center, Araras, São Paulo, Brazil
| | - Milton Santamaria-Jr
- Graduate Program of Biomedical Sciences, Herminio Ometto University Center, Araras, São Paulo, Brazil.,Graduate Program of Orthodontics, Herminio Ometto University Center, Araras, São Paulo, Brazil
| |
Collapse
|
23
|
Chen G, Deng S, Zuo M, Wang J, Cheng D, Chen B. Non-viral CRISPR activation system targeting VEGF-A and TGF-β1 for enhanced osteogenesis of pre-osteoblasts implanted with dual-crosslinked hydrogel. Mater Today Bio 2022; 16:100356. [PMID: 35898441 PMCID: PMC9309523 DOI: 10.1016/j.mtbio.2022.100356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
Healing of large calvarial bone defects remains challenge but may be improved by stimulating bone regeneration of implanted cells. The aim of this study is to specially co-activate transforming growth factor β1 (TGF-β1) and vascular endothelial growth factor (VEGF-A) genes expressions in pre-osteoblast MC3T3-E1 cells through the non-viral CRISPR activation (CRISPRa) system to promote osteogenesis. A cationic copolymer carrying nucleus localizing peptides and proton sponge groups dimethyl-histidine was synthesized to deliver CRISPRa system into MC3T3-E1 cells with high cellular uptake, lysosomal escape, and nuclear translocation, which activated VEGF-A and TGF-β1 genes expressions and thereby additively or synergistically induced several osteogenic genes expressions. A tunable dual-crosslinked hydrogel was developed to implant the above engineered cells into mice calvaria bone defect site to promote bone healing in vivo. The combination of multi-genes activation through non-viral CRISPRa system and tunable dual-crosslinked hydrogel provides a versatile strategy for promoting bone healing with synergistic effect.
Collapse
Affiliation(s)
- Guo Chen
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, PR China
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Shaohui Deng
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Mingxiang Zuo
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Jin Wang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
| | - Du Cheng
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, PR China
- Corresponding author.
| | - Bin Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Corresponding author.
| |
Collapse
|
24
|
Deletion of osteopontin or bone sialoprotein induces opposite bone responses to mechanical stimulation in mice. Bone Rep 2022; 17:101621. [PMID: 36159882 PMCID: PMC9493388 DOI: 10.1016/j.bonr.2022.101621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/21/2022] Open
Abstract
Osteopontin (OPN) and Bone Sialoprotein (BSP) are co-expressed in bone and display overlapping and complementary physiological properties. Both genes show a rapid expression response to mechanical stimulation. We used mice with single and double deletions (DKO) of BSP and OPN to assess the specificity of their roles in skeletal adaptation to loading. Two-month-old Wild-Type (WT), BSP knockout (BSP−/−), OPN−/− and DKO male mice were submitted to two mechanical stimulation regimen (n = 10 mice/group) respectively impacting trabecular bone (Hypergravity, HG) and cortical bone (Whole Body Vibration, WBV). HG increased trabecular bone volume (BV/TV) in WT femur through reduced resorption, and in BSP−/− mice femur and vertebra through increased bone formation. In contrast, HG increased the turnover of OPN−/− bone, resulting in reduced femur and vertebra BV/TV. HG did not affect DKO bones. Similarly, WBV increased cortical thickness in BSP−/− mice and decreased it in OPN−/−, without affecting structurally WT and DKO bone. Vibrated BSP−/− mice displayed increased endocortical bone formation with a drop in Sclerostin expression, and reduced periosteal osteoclasts with lower Rankl and Cathepsin K expression. In contrast, vibrated OPN−/− endocortical bone displayed decreased formation and increased osteoclast coverage. Therefore, under two regimen (HG and WBV) targeting distinct bone compartments, absence of OPN resulted in bone loss while lack of BSP induced bone gain, reflecting divergent structural adaptations. Strikingly, absence of both proteins led to a relative insensitivity to either mechanical challenge. Interplay between OPN and BSP thus appears as a key element of skeletal response to mechanical stimulation. Osteopontin gene knockout induces bone loss under mechanical stimulation Bone Sialoprotein gene knockout potentiates bone gain under mechanical stimulation Knockout of both genes leads to bone insensitivity Their interplay is crucial for bone response to mechanical challenges
Collapse
|
25
|
Nagy E, Sobh MM, Abdalbary M, Elnagar S, Elrefaey R, Shabaka S, Elshabrawy N, Shemies R, Tawfik M, Santos CGS, Barreto FC, El-Husseini A. Is Adynamic Bone Always a Disease? Lessons from Patients with Chronic Kidney Disease. J Clin Med 2022; 11:jcm11237130. [PMID: 36498703 PMCID: PMC9736225 DOI: 10.3390/jcm11237130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Renal osteodystrophy (ROD) is a common complication of end-stage kidney disease that often starts early with loss of kidney function, and it is considered an integral part in management of patients with chronic kidney disease (CKD). Adynamic bone (ADB) is characterized by suppressed bone formation, low cellularity, and thin osteoid seams. There is accumulating evidence supporting increasing prevalence of ADB, particularly in early CKD. Contemporarily, it is not very clear whether it represents a true disease, an adaptive mechanism to prevent bone resorption, or just a transitional stage. Several co-players are incriminated in its pathogenesis, such as age, diabetes mellitus, malnutrition, uremic milieu, and iatrogenic factors. In the present review, we will discuss the up-to-date knowledge of the ADB and focus on its impact on bone health, fracture risk, vascular calcification, and long-term survival. Moreover, we will emphasize the proper preventive and management strategies of ADB that are pivotal issues in managing patients with CKD. It is still unclear whether ADB is always a pathologic condition or whether it can represent an adaptive process to suppress bone resorption and further bone loss. In this article, we tried to discuss this hard topic based on the available limited information in patients with CKD. More studies are needed to be able to clearly address this frequent ROD finding.
Collapse
Affiliation(s)
- Eman Nagy
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura 35516, Egypt
| | - Mahmoud M. Sobh
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed Abdalbary
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura 35516, Egypt
| | - Sherouk Elnagar
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura 35516, Egypt
| | - Rabab Elrefaey
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura 35516, Egypt
| | - Shimaa Shabaka
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura 35516, Egypt
| | - Nehal Elshabrawy
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura 35516, Egypt
| | - Rasha Shemies
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura 35516, Egypt
| | - Mona Tawfik
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura 35516, Egypt
| | - Cássia Gomes S. Santos
- Department of Internal Medicine, Division of Nephrology, Federal University of Paraná, Curitiba 80060-00, PR, Brazil
| | - Fellype C. Barreto
- Department of Internal Medicine, Division of Nephrology, Federal University of Paraná, Curitiba 80060-00, PR, Brazil
| | - Amr El-Husseini
- Division of Nephrology & Bone and Mineral Metabolism, University of Kentucky, Lexington, KY 40536-0298, USA
- Correspondence: ; Tel.: +1-859-218-0934; Fax: +1-859-323-0232
| |
Collapse
|
26
|
Associated changes in stiffness of collagen scaffolds during osteoblast mineralisation and bone formation. BMC Res Notes 2022; 15:310. [PMID: 36153566 PMCID: PMC9509582 DOI: 10.1186/s13104-022-06203-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/07/2022] [Indexed: 11/10/2022] Open
Abstract
Objective Engineering bone in 3D is important for both regenerative medicine purposes and for the development of accurate in vitro models of bone tissue. The changing material stiffness of bone tissue had not yet been monitored throughout the process of mineralisation and bone nodule formation by osteoblasts either during in vitro engineering or in development perspective. Results Within this short research note, stiffness changes (Young’s modulus) during in vitro bone formation by primary osteoblasts in dense collagen scaffolds were monitored using atomic force microscopy. Data analysis revealed significant stiffening of 3D bone cultures at day 5 and 8 that was correlated with the onset of mineral deposition (p < 0.00005).
Collapse
|
27
|
Hasegawa T, Hongo H, Yamamoto T, Abe M, Yoshino H, Haraguchi-Kitakamae M, Ishizu H, Shimizu T, Iwasaki N, Amizuka N. Matrix Vesicle-Mediated Mineralization and Osteocytic Regulation of Bone Mineralization. Int J Mol Sci 2022; 23:ijms23179941. [PMID: 36077336 PMCID: PMC9456179 DOI: 10.3390/ijms23179941] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Bone mineralization entails two mineralization phases: primary and secondary mineralization. Primary mineralization is achieved when matrix vesicles are secreted by osteoblasts, and thereafter, bone mineral density gradually increases during secondary mineralization. Nearby extracellular phosphate ions (PO43−) flow into the vesicles via membrane transporters and enzymes located on the vesicles’ membranes, while calcium ions (Ca2+), abundant in the tissue fluid, are also transported into the vesicles. The accumulation of Ca2+ and PO43− in the matrix vesicles induces crystal nucleation and growth. The calcium phosphate crystals grow radially within the vesicle, penetrate the vesicle’s membrane, and continue to grow outside the vesicle, ultimately forming mineralized nodules. The mineralized nodules then attach to collagen fibrils, mineralizing them from the contact sites (i.e., collagen mineralization). Afterward, the bone mineral density gradually increases during the secondary mineralization process. The mechanisms of this phenomenon remain unclear, but osteocytes may play a key role; it is assumed that osteocytes enable the transport of Ca2+ and PO43− through the canaliculi of the osteocyte network, as well as regulate the mineralization of the surrounding bone matrix via the Phex/SIBLINGs axis. Thus, bone mineralization is biologically regulated by osteoblasts and osteocytes.
Collapse
Affiliation(s)
- Tomoka Hasegawa
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
- Correspondence: (T.H.); (N.A.); Tel.: +81-11-706-4226 (T.H.); +81-11-706-4223 (N.A.)
| | - Hiromi Hongo
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| | - Tomomaya Yamamoto
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
- Northern Army Medical Unit, Camp Makomanai, Japan Ground Self-Defense Forces, Sapporo 005-8543, Japan
| | - Miki Abe
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| | - Hirona Yoshino
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| | - Mai Haraguchi-Kitakamae
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
- Division of Craniofacial Development and Tissue Biology, Graduate School of Dentistry, Tohoku University, Sendai 980-8577, Japan
| | - Hotaka Ishizu
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
- Orthopedic Surgery, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Tomohiro Shimizu
- Orthopedic Surgery, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Norimasa Iwasaki
- Orthopedic Surgery, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Norio Amizuka
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
- Correspondence: (T.H.); (N.A.); Tel.: +81-11-706-4226 (T.H.); +81-11-706-4223 (N.A.)
| |
Collapse
|
28
|
Figueredo CA, Abdelhay N, Ganatra S, Gibson MP. The role of Dentin Sialophosphoprotein (DSPP) in craniofacial development. J Oral Biol Craniofac Res 2022; 12:673-678. [DOI: 10.1016/j.jobcr.2022.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/17/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
|
29
|
Lorencetti-Silva F, Sales LS, Lamarque GDCC, Caixeta GA, Arnez MFM, Faccioli LH, Paula-Silva FWG. Effects of inflammation in dental pulp cell differentiation and reparative response. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.942714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The responsiveness of the dentin-pulp complex is possible due to the stimulation of dental pulp cells, which begin to synthesize and secrete dentin matrix. The inflammatory process generated by harmful stimuli should be understood as a natural event of the immune response, resulting in the recruitment of hematopoietic cells, which cross the endothelial barrier and reach the site affected by the injury in order to eliminate the damage and provide an appropriate environment for the restoration of homeostasis. The repair process occurs in the presence of adequate blood supply, absence of infection, and with the participation of pro-inflammatory cytokines, growth factors, extracellular matrix components, and other biologically active molecules. Prostaglandins and leukotrienes are bioactive molecules derived from the metabolism of arachidonic acid, as a result of a variable range of cellular stimuli. The aim of this review is to describe the process of formation and biomineralization of the dentin-pulp complex and how pro-inflammatory events can modify this response, with emphasis on the lipid mediators prostaglandins and leukotrienes derived from arachidonic acid metabolism.
Collapse
|
30
|
Li Z, Cong X, Kong W. Matricellular proteins: Potential biomarkers and mechanistic factors in aortic aneurysms. J Mol Cell Cardiol 2022; 169:41-56. [DOI: 10.1016/j.yjmcc.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/30/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
31
|
Wang Q, Wang H, Ding Y, Wan M, Xu M. The Role of Adipokines in Pancreatic Cancer. Front Oncol 2022; 12:926230. [PMID: 35875143 PMCID: PMC9305334 DOI: 10.3389/fonc.2022.926230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
In modern society, inappropriate diets and other lifestyle habits have made obesity an increasingly prominent health problem. Pancreatic cancer (PC), a kind of highly aggressive malignant tumor, is known as a silent assassin and is the seventh leading cause of cancer death worldwide, pushing modern medicine beyond help. Adipokines are coming into notice because of the role of the intermediate regulatory junctions between obesity and malignancy. This review summarizes the current evidence for the relationship between highly concerning adipokines and the pathogenesis of PC. Not only are classical adipokines such as leptin and adiponectin included, but they also cover the recognized chemerin and osteopontin. Through a summary of the biological functions of these adipokines as well as their receptors, it was discovered that in addition to their basic function of stimulating the biological activity of tumors, more studies confirm that adipokines intervene in the progression of PC from the viewpoint of tumor metabolism, immune escape, and reprogramming of the tumor microenvironment (TME). Besides endocrine function, the impact of white adipose tissue (WAT)-induced chronic inflammation on PC is briefly discussed. Furthermore, the potential implication of the acknowledged endocrine behavior of brown adipose tissue (BAT) in relation to carcinogenesis is also explored. No matter the broad spectrum of obesity and the poor prognosis of PC, supplemental research is needed to unravel the detailed network of adipokines associated with PC. Exploiting profound therapeutic strategies that target adipokines and their receptors may go some way to improving the current worrying prognosis of PC patients.
Collapse
|
32
|
Figueredo CA, Abdelhay N, Gibson MP. The Roles of SIBLING Proteins in Dental, Periodontal and Craniofacial Development. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.898802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The majority of dental, periodontal, and craniofacial tissues are derived from the neural crest cells and ectoderm. Neural crest stem cells are pluripotent, capable of differentiating into a variety of cells. These cells can include osteoblasts, odontoblasts, cementoblasts, chondroblasts, and fibroblasts which are responsible for forming some of the tissues of the oral and craniofacial complex. The hard tissue forming cells deposit a matrix composed of collagen and non-collagenous proteins (NCPs) that later undergoes mineralization. The NCPs play a role in the mineralization of collagen. One such category of NCPs is the small integrin-binding ligand, N-linked glycoprotein (SIBLING) family of proteins. This family is composed of dentin sialophosphosprotein (DSPP), osteopontin (OPN), dentin matrix protein 1 (DMP1), bone sialoprotein (BSP), and matrix extracellular phosphoglycoprotein (MEPE). The SIBLING family is known to have regulatory effects in the mineralization process of collagen fibers and the maturation of hydroxyapatite crystals. It is well established that SIBLING proteins have critical roles in tooth development. Recent literature has described the expression and role of SIBLING proteins in other areas of the oral and craniofacial complex as well. The objective of the present literature review is to summarize and discuss the different roles the SIBLING proteins play in the development of dental, periodontal, and craniofacial tissues.
Collapse
|
33
|
Tian C, Chai J, Liu W, Zhang X, Li Y, Zuo H, Yuan G, Zhang H, Liu H, Chen Z. Role of the Demethylase AlkB Homolog H5 in the Promotion of Dentinogenesis. Front Physiol 2022; 13:923185. [PMID: 35784864 PMCID: PMC9240783 DOI: 10.3389/fphys.2022.923185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022] Open
Abstract
Dentinogenesis is a key process in tooth formation and is regulated by a series of pre- and post-transcriptional regulations. N6-methyl-adenosine (m6A), which is the most prevalent internal chemical modification that can be removed by the RNA demethylase AlkB homolog H5 (ALKBH5), has recently been reported to be involved in several biological processes. However, the exact function of ALKBH5-mediated m6A modification in tooth development remains unclear. Here, we showed that Alkbh5 was expressed in pre-odontoblasts, polarizing odontoblasts, and secretory odontoblasts. Alkbh5 overexpression in the mouse dental papilla cell line mDPC6T promoted odontoblastic differentiation. Conditional knockout of Alkbh5 in Dmp1-expressing odontoblasts led to a decrease in number of odontoblasts and increased pre-dentin formation. Mechanistically, RNA sequencing and m6A sequencing of Alkbh5-overexpressing mDPC6T cells revealed that Alkbh5 promoted odontoblast differentiation by prolonging the half-life of Runx2 transcripts in an m6A-dependent manner and by activating the phosphatidylinositol 3-kinase/protein kinase B pathway. Notably, the loss of Alkbh5 expression in odontoblasts impaired tertiary dentin formation in vivo. These results suggested that the RNA demethylase ALKBH5 plays a role in dentinogenesis.
Collapse
Affiliation(s)
- Cheng Tian
- The State Key Laboratory Breeding Base of Basic Sciences of Stomatology, Key Laboratory of Oral Biomedicine, Ministry of Education (Hubei-MOST KLOS & KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jihua Chai
- The State Key Laboratory Breeding Base of Basic Sciences of Stomatology, Key Laboratory of Oral Biomedicine, Ministry of Education (Hubei-MOST KLOS & KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Weidong Liu
- The State Key Laboratory Breeding Base of Basic Sciences of Stomatology, Key Laboratory of Oral Biomedicine, Ministry of Education (Hubei-MOST KLOS & KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xinye Zhang
- The State Key Laboratory Breeding Base of Basic Sciences of Stomatology, Key Laboratory of Oral Biomedicine, Ministry of Education (Hubei-MOST KLOS & KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yashu Li
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Huanyan Zuo
- The State Key Laboratory Breeding Base of Basic Sciences of Stomatology, Key Laboratory of Oral Biomedicine, Ministry of Education (Hubei-MOST KLOS & KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guohua Yuan
- The State Key Laboratory Breeding Base of Basic Sciences of Stomatology, Key Laboratory of Oral Biomedicine, Ministry of Education (Hubei-MOST KLOS & KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Haojian Zhang
- The State Key Laboratory Breeding Base of Basic Sciences of Stomatology, Key Laboratory of Oral Biomedicine, Ministry of Education (Hubei-MOST KLOS & KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Huan Liu
- The State Key Laboratory Breeding Base of Basic Sciences of Stomatology, Key Laboratory of Oral Biomedicine, Ministry of Education (Hubei-MOST KLOS & KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- *Correspondence: Huan Liu, ; Zhi Chen,
| | - Zhi Chen
- The State Key Laboratory Breeding Base of Basic Sciences of Stomatology, Key Laboratory of Oral Biomedicine, Ministry of Education (Hubei-MOST KLOS & KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- *Correspondence: Huan Liu, ; Zhi Chen,
| |
Collapse
|
34
|
Xiao L, Xu Q, Liu X, Chan S, Luo Y, He S, Fang M. The Novel-miR-659/SPP1 Interaction Regulates Fat Deposition in Castrated Male Pigs. Animals (Basel) 2022; 12:ani12080944. [PMID: 35454191 PMCID: PMC9031235 DOI: 10.3390/ani12080944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/20/2022] [Accepted: 03/31/2022] [Indexed: 11/26/2022] Open
Abstract
Simple Summary Castration is a standard method for eliminating boar taint in industrial hog production, but it also causes enormous fat accumulation in the carcass. Secreted phosphoprotein 1 (SPP1) was selected to investigate its functions on the regulation of adipose deposition based on our previous data. In the present study, SPP1 overexpression and interference bidirectionally verified that SPP1 inhibited adipogenic differentiation of porcine bone marrow mesenchymal stem cells (pBMSCs). Testosterone-treated cell models were used to simulate the androgen status of intact pigs, and testosterone addition influenced SPP1 mRNA levels during the differentiation of pBMSCs. Moreover, we identified novel-miR-659 and targeted the 3′ untranslated region of SPP1 based on bioinformatics analysis and dual-luciferase assays, and found that the novel-miR-659 upregulation promoted adipogenesis while novel-miR-659 downregulation suppressed adipogenesis in pBMSCs detected by Oil Red O staining and adipogenic markers. Collectively, the interaction between novel-miR-659 and SPP1 can regulate adipose accumulation in castrated male pigs. Our data provide a theoretical basis for further study on the fat deposition mechanism caused by castration. Abstract Castration is usually used to remove boar taint in commercial pork production, but the adipose accumulation was increased excessively, which affected the meat quality of pigs. Based on our previous study, secreted phosphoprotein 1 (SPP1) was significantly differentially expressed between castrated and intact male pigs. However, the role of SPP1 in regulating adipose growth and fat storage caused by castration is unknown. In this study, SPP1 was identified to inhibit adipogenesis by the expression of adipogenic markers PPARγ and FABP4 as well as Oil red staining assay during differentiation of porcine bone marrow mesenchymal stem cells (pBMSCs). Subsequently, testosterone was used to treat pBMSCs to simulate the androgen status of intact pigs. Compared with the control groups without testosterone, the SPP1 expression in the testosterone group was markedly increased in the late stage of pBMSCs differentiation. Furthermore, novel-miR-659 was predicted by TargetScan and miRDB to target SPP1 and verified through a dual-luciferase reporter assay. Oil Red O staining assay indicated that novel-miR-659 overexpression significantly promoted adipogenesis, whereas novel-miR-659 inhibition suppressed adipogenesis. The expressions of adipogenic markers PPARγ and FABP4 showed the same tendency. Taken together, our study found that the targeted interaction between novel-miR-659 and SPP1 is involved in regulation of fat deposition in castrated male pigs.
Collapse
Affiliation(s)
- Lianmei Xiao
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (L.X.); (Q.X.); (X.L.); (S.C.); (Y.L.); (S.H.)
| | - Qiao Xu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (L.X.); (Q.X.); (X.L.); (S.C.); (Y.L.); (S.H.)
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ximing Liu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (L.X.); (Q.X.); (X.L.); (S.C.); (Y.L.); (S.H.)
| | - Shuheng Chan
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (L.X.); (Q.X.); (X.L.); (S.C.); (Y.L.); (S.H.)
| | - Yabiao Luo
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (L.X.); (Q.X.); (X.L.); (S.C.); (Y.L.); (S.H.)
| | - Shuaihan He
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (L.X.); (Q.X.); (X.L.); (S.C.); (Y.L.); (S.H.)
| | - Meiying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (L.X.); (Q.X.); (X.L.); (S.C.); (Y.L.); (S.H.)
- Sanya Institute of China Agricultural University, Sanya 572025, China
- Correspondence: ; Tel./Fax: +86-10-62734943
| |
Collapse
|
35
|
Leukotriene B 4 loaded in microspheres regulate the expression of genes related to odontoblastic differentiation and biomineralization by dental pulp stem cells. BMC Oral Health 2022; 22:45. [PMID: 35197043 PMCID: PMC8864908 DOI: 10.1186/s12903-022-02083-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/14/2022] [Indexed: 11/29/2022] Open
Abstract
Background Leukotriene B4 (LTB4) is a potent lipid mediator that stimulate the immune response. Because dental pulp inflammation and dentin repair are intrinsically related responses, the aim of this research was to investigate the potential of LTB4 in inducing differentiation of dental pulp stem cells.
Methods Microspheres (MS) loaded with LTB4 were prepared using an oil emulsion solvent extraction evaporation process and sterility, characterization, efficiency of LTB4 encapsulation and in vitro LTB4 release assay were investigated. Mouse dental pulp stem cells (OD-21) were stimulated with soluble LTB4 or MS loaded with LTB4 (0.01 and 0.1 μM). Cytotoxicity and cell viability was determined by lactate dehydrogenase and methylthiazol tetrazolium assays. Gene expression were measured by quantitative reverse transcription polymerase chain reaction after 3, 6, 24, 48 and 72 h. Mineralized nodule formation was assessed after 28 days of OD-21 cell stimulation with LTB4 in mineralized media or not. Groups were compared using one-way ANOVA test followed by Dunnett’s post-test (α = 0.05).
Results Treatment with LTB4 or MS loaded with LTB4 (0.01 and 0.1 µm-μM) were not cytotoxic to OD-21 cells. Treatment with LTB4 modulated the expression of the Ibsp (integrin binding sialoprotein) and Runx2 (runt-related transcription factor 2) genes differently depending on the experimental period analyzed. Interestingly LTB4 loaded in microspheres (0.1 μM) allowed long term dental pulp cell differentiation and biomineralization. Conclusion LTB4, soluble or loaded in MS, were not cytotoxic and modulated the expression of the Ibsp and Runx2 genes in cultured OD-21 cells. When LTB4 was incorporated into MS, odontoblast differentiation and mineralization was induced in long term culture.
Collapse
|
36
|
Bioengineering the ameloblastoma tumour to study its effect on bone nodule formation. Sci Rep 2021; 11:24088. [PMID: 34916549 PMCID: PMC8677805 DOI: 10.1038/s41598-021-03484-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/03/2021] [Indexed: 01/09/2023] Open
Abstract
Ameloblastoma is a benign, epithelial cancer of the jawbone, which causes bone resorption and disfigurement to patients affected. The interaction of ameloblastoma with its tumour stroma drives invasion and progression. We used stiff collagen matrices to engineer active bone forming stroma, to probe the interaction of ameloblastoma with its native tumour bone microenvironment. This bone-stroma was assessed by nano-CT, transmission electron microscopy (TEM), Raman spectroscopy and gene analysis. Furthermore, we investigated gene correlation between bone forming 3D bone stroma and ameloblastoma introduced 3D bone stroma. Ameloblastoma cells increased expression of MMP-2 and -9 and RANK temporally in 3D compared to 2D. Our 3D biomimetic model formed bone nodules of an average surface area of 0.1 mm2 and average height of 92.37 [Formula: see text] 7.96 μm over 21 days. We demonstrate a woven bone phenotype with distinct mineral and matrix components and increased expression of bone formation genes in our engineered bone. Introducing ameloblastoma to the bone stroma, completely inhibited bone formation, in a spatially specific manner. Multivariate gene analysis showed that ameloblastoma cells downregulate bone formation genes such as RUNX2. Through the development of a comprehensive bone stroma, we show that an ameloblastoma tumour mass prevents osteoblasts from forming new bone nodules and severely restricted the growth of existing bone nodules. We have identified potential pathways for this inhibition. More critically, we present novel findings on the interaction of stromal osteoblasts with ameloblastoma.
Collapse
|
37
|
Enzymatic Approach in Calcium Phosphate Biomineralization: A Contribution to Reconcile the Physicochemical with the Physiological View. Int J Mol Sci 2021; 22:ijms222312957. [PMID: 34884758 PMCID: PMC8657759 DOI: 10.3390/ijms222312957] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 01/22/2023] Open
Abstract
Biomineralization is the process by which organisms produce hard inorganic matter from soft tissues with outstanding control of mineral deposition in time and space. For this purpose, organisms deploy a sophisticated "toolkit" that has resulted in significant evolutionary innovations, for which calcium phosphate (CaP) is the biomineral selected for the skeleton of vertebrates. While CaP mineral formation in aqueous media can be investigated by studying thermodynamics and kinetics of phase transitions in supersaturated solutions, biogenic mineralization requires coping with the inherent complexity of biological systems. This mainly includes compartmentalization and homeostatic processes used by organisms to regulate key physiological factors, including temperature, pH and ion concentration. A detailed analysis of the literature shows the emergence of two main views describing the mechanism of CaP biomineralization. The first one, more dedicated to the study of in vivo systems and supported by researchers in physiology, often involves matrix vesicles (MVs). The second one, more investigated by the physicochemistry community, involves collagen intrafibrillar mineralization particularly through in vitro acellular models. Herein, we show that there is an obvious need in the biological systems to control both where and when the mineral forms through an in-depth survey of the mechanism of CaP mineralization. This necessity could gather both communities of physiologists and physicochemists under a common interest for an enzymatic approach to better describe CaP biomineralization. Both homogeneous and heterogeneous enzymatic catalyses are conceivable for these systems, and a few preliminary promising results on CaP mineralization for both types of enzymatic catalysis are reported in this work. Through them, we aim to describe the relevance of our point of view and the likely findings that could be obtained when adding an enzymatic approach to the already rich and creative research field dealing with CaP mineralization. This complementary approach could lead to a better understanding of the biomineralization mechanism and inspire the biomimetic design of new materials.
Collapse
|
38
|
Shen J, Zhao M, Zhang C, Sun X. IL-1β in atherosclerotic vascular calcification: From bench to bedside. Int J Biol Sci 2021; 17:4353-4364. [PMID: 34803503 PMCID: PMC8579452 DOI: 10.7150/ijbs.66537] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/11/2021] [Indexed: 01/19/2023] Open
Abstract
Atherosclerotic vascular calcification contributes to increased risk of death in patients with cardiovascular diseases. Assessing the type and severity of inflammation is crucial in the treatment of numerous cardiovascular conditions. IL-1β, a potent proinflammatory cytokine, plays diverse roles in the pathogenesis of atherosclerotic vascular calcification. Several large-scale, population cohort trials have shown that the incidence of cardiovascular events is clinically reduced by the administration of anti-IL-1β therapy. Anti-IL-1β therapy might reduce the incidence of cardiovascular events by affecting atherosclerotic vascular calcification, but the mechanism underlying this effect remains unclear. In this review, we summarize current knowledge on the role of IL-1β in atherosclerotic vascular calcification, and describe the latest results reported in clinical trials evaluating anti-IL-1β therapies for the treatment of cardiovascular diseases. This review will aid in improving current understanding of the pathophysiological roles of IL-1β and mechanisms underlying its activity.
Collapse
Affiliation(s)
- Jialing Shen
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ming Zhao
- Department of Interventional Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Chunxiang Zhang
- Laboratory of Nucleic Acids in Medicine for National high-level talents, Southwest Medical University, Luzhou 646000, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Xiaolei Sun
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Department of Interventional Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Laboratory of Nucleic Acids in Medicine for National high-level talents, Southwest Medical University, Luzhou 646000, China.,School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Research Excellence, Faculty of Life Science and Medicine, King's College London, London SE5 9NU, United Kingdom.,Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, China
| |
Collapse
|
39
|
Abstract
Biomineralization of enamel, dentin, and bone involves the deposition of apatite mineral crystals within an organic matrix. Bone and teeth are classic examples of biomaterials with unique biomechanical properties that are crucial to their function. The collagen-based apatite mineralization and the important function of noncollagenous proteins are similar in dentin and bone; however, enamel is formed in a unique amelogenin-containing protein matrix. While the structure and organic composition of enamel are different from those of dentin and bone, the principal molecular mechanisms of protein-protein interactions, protein self-assembly, and control of crystallization events by the organic matrix are common among these apatite-containing tissues. This review briefly summarizes enamel and dentin matrix components and their interactions with other extracellular matrix components and calcium ions in mediating the mineralization process. We highlight the crystallization events that are controlled by the protein matrix and their interactions in the extracellular matrix during enamel and dentin biomineralization. Strategies for peptide-inspired biomimetic growth of tooth enamel and bioinspired mineralization of collagen to stimulate repair of demineralized dentin and bone tissue engineering are also addressed.
Collapse
Affiliation(s)
- J Moradian-Oldak
- Center for Craniofacial Molecular Biology, Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - A George
- Brodie Tooth Development Genetics & Regenerative Medicine Research Laboratory, Department of Oral Biology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
40
|
Shaping collagen for engineering hard tissues: Towards a printomics approach. Acta Biomater 2021; 131:41-61. [PMID: 34192571 DOI: 10.1016/j.actbio.2021.06.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022]
Abstract
Hard tissue engineering has evolved over the past decades, with multiple approaches being explored and developed. Despite the rapid development and success of advanced 3D cell culture, 3D printing technologies and material developments, a gold standard approach to engineering and regenerating hard tissue substitutes such as bone, dentin and cementum, has not yet been realised. One such strategy that differs from conventional regenerative medicine approach of other tissues, is the in vitro mineralisation of collagen templates in the absence of cells. Collagen is the most abundant protein within the human body and forms the basis of all hard tissues. Once mineralised, collagen provides important support and protection to humans, for example in the case of bone tissue. Multiple in vitro fabrication strategies and mineralisation approaches have been developed and their success in facilitating mineral deposition on collagen to achieve bone-like scaffolds evaluated. Critical to the success of such fabrication and biomineralisation approaches is the collagen template, and its chemical composition, organisation, and density. The key factors that influence such properties are the collagen processing and fabrication techniques utilised to create the template, and the mineralisation strategy employed to deposit mineral on and throughout the templates. However, despite its importance, relatively little attention has been placed on these two critical factors. Here, we critically examine the processing, fabrication and mineralisation strategies that have been used to mineralise collagen templates, and offer insights and perspectives on the most promising strategies for creating mineralised collagen scaffolds. STATEMENT OF SIGNIFICANCE: In this review, we highlight the critical need to fabricate collagen templates with advanced processing techniques, in a manner that achieves biomimicry of the hierarchical collagen structure, prior to utilising in vitro mineralisation strategies. To this end, we focus on the initial collagen that is selected, the extraction techniques used and the native fibril forming potential retained to create reconstituted collagen scaffolds. This review synthesises current best practises in material sourcing, processing, mineralisation strategies and fabrication techniques, and offers insights into how these can best be exploited in future studies to successfully mineralise collagen templates.
Collapse
|
41
|
Yan T, Kong Y, Fan W, Kang J, Chen H, He H, Huang F. Expression of nitric oxide synthases in rat odontoblasts and the role of nitric oxide in odontoblastic differentiation of rat dental papilla cells. Dev Growth Differ 2021; 63:354-371. [PMID: 34411285 DOI: 10.1111/dgd.12745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/23/2021] [Accepted: 08/08/2021] [Indexed: 12/01/2022]
Abstract
As precursor cells of odontoblasts, dental papilla cells (DPCs) form the dentin-pulp complex during tooth development. Nitric oxide (NO) regulates the functions of multiple cells and organ tissues, including stem cell differentiation and bone formation. In this paper, we explored the involvement of NO in odontoblastic differentiation. We verified the expression of NO synthase (NOS) in rat odontoblasts by nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) staining and immunohistochemistry in vivo. The expression of all three NOS isoforms in rat DPCs was confirmed by quantitative reverse-transcription polymerase chain reaction (qRT-PCR), immunofluorescence, and western blotting in vitro. The expression of neuronal NOS and endothelial NOS was upregulated during the odontoblastic differentiation of DPCs. Inhibition of NOS function by NOS inhibitor l-NG -monomethyl arginine (L-NMMA) resulted in reduced formation of mineralized nodules and expression of dentin sialophosphoprotein (DSPP) and dentin matrix protein (DMP1) during DPC differentiation. The NO donor S-nitroso-N-acetylpenicillamine (SNAP, 0.1, 1, 10, and 100 μM) promoted the viability of DPCs. Extracellular matrix mineralization and odontogenic markers expression were elevated by SNAP at low concentrations (0.1, 1, and 10 μM) and suppressed at high concentration (100 μM). Blocking the generation of cyclic guanosine monophosphate (cGMP) with 1H-(1,2,4)oxadiazolo-(4,3-a)quinoxalin-1-one (ODQ) abolished the positive influence of SNAP on the odontoblastic differentiation of DPCs. These findings demonstrate that NO regulates the odontoblastic differentiation of DPCs, thereby influencing dentin formation and tooth development.
Collapse
Affiliation(s)
- Tong Yan
- Department of Pediatric Dentistry, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yu Kong
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenguo Fan
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jun Kang
- Department of Pediatric Dentistry, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Haoling Chen
- Department of Pediatric Dentistry, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Fang Huang
- Department of Pediatric Dentistry, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
42
|
Luo X, Yin J, Miao S, Feng W, Ning T, Xu S, Huang S, Zhang S, Liao Y, Hao C, Wu B, Ma D. mTORC1 promotes mineralization via p53 pathway. FASEB J 2021; 35:e21325. [PMID: 33508145 DOI: 10.1096/fj.202002016r] [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: 09/02/2020] [Revised: 12/04/2020] [Accepted: 12/14/2020] [Indexed: 12/30/2022]
Abstract
The objectives of our study were to investigate the roles of mTORC1 in odontoblast proliferation and mineralization and to determine the mechanism by which mTORC1 regulates odontoblast mineralization. In vitro, MDPC23 cells were treated with rapamycin (10 nmol/L) and transfected with a lentivirus for short hairpin (shRNA)-mediated silencing of the tuberous sclerosis complex (shTSC1) to inhibit and activate mTORC1, respectively. CCK8 assays, flow cytometry, Alizarin red S staining, ALP staining, qRT-PCR, and western blot analysis were performed. TSC1-conditional knockout (DMP1-Cre+ ; TSC1f/f , hereafter CKO) mice and littermate control (DMP1-Cre- ; TSC1f/f , hereafter WT) mice were generated. H&E staining, immunofluorescence, and micro-CT analysis were performed. Transcriptome sequencing analysis was used to screen the mechanism of this process. mTORC1 inactivation decreased the cell proliferation. The qRT-PCR and western blot results showed that mineralization-related genes and proteins were downregulated in mTORC1-inactivated cells. Moreover, mTORC1 overactivation promoted cell proliferation and mineralization-related gene and protein expression. In vivo, the micro-CT results showed that DV/TV and dentin thickness were higher in CKO mice than in controls and H&E staining showed the same results. Mineralization-related proteins expression was upregulated. Transcriptome sequencing analysis revealed that p53 pathway-associated genes were differentially expressed in TSC1-deficient cells. By inhibiting p53 alone or both mTORC1 and p53 with rapamycin and a p53 inhibitor, we elucidated that p53 acts downstream of mTORC1 and that mTORC1 thereby promotes odontoblast mineralization. Taken together, our findings demonstrate that the role of mTORC1 in odontoblast proliferation and mineralization, and confirm that mTORC1 upregulates odontoblast mineralization via the p53 pathway.
Collapse
Affiliation(s)
- Xinghong Luo
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,College of Stomatology, Southern Medical University, Guangzhou, China
| | - Jingyao Yin
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,College of Stomatology, Southern Medical University, Guangzhou, China
| | - Shenghong Miao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,College of Stomatology, Southern Medical University, Guangzhou, China
| | - Weiqing Feng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,College of Stomatology, Southern Medical University, Guangzhou, China
| | - Tingting Ning
- College of Stomatology, Southern Medical University, Guangzhou, China.,Department of Endodontics, Stomatology Hospital, Southern Medical University, Guangzhou, China
| | - Shuaimei Xu
- College of Stomatology, Southern Medical University, Guangzhou, China.,Department of Endodontics, Stomatology Hospital, Southern Medical University, Guangzhou, China
| | - Shijiang Huang
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Sheng Zhang
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunbo Hao
- Department of Stomatology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Buling Wu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,College of Stomatology, Southern Medical University, Guangzhou, China
| | - Dandan Ma
- Department of Endodontics, Stomatology Hospital, Southern Medical University, Guangzhou, China.,Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, USA
| |
Collapse
|
43
|
Nikoloudaki G. Functions of Matricellular Proteins in Dental Tissues and Their Emerging Roles in Orofacial Tissue Development, Maintenance, and Disease. Int J Mol Sci 2021; 22:ijms22126626. [PMID: 34205668 PMCID: PMC8235165 DOI: 10.3390/ijms22126626] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/04/2023] Open
Abstract
Matricellular proteins (MCPs) are defined as extracellular matrix (ECM) associated proteins that are important regulators and integrators of microenvironmental signals, contributing to the dynamic nature of ECM signalling. There is a growing understanding of the role of matricellular proteins in cellular processes governing tissue development as well as in disease pathogenesis. In this review, the expression and functions of different MP family members (periostin, CCNs, TSPs, SIBLINGs and others) are presented, specifically in relation to craniofacial development and the maintenance of orofacial tissues, including bone, gingiva, oral mucosa, palate and the dental pulp. As will be discussed, each MP family member has been shown to have non-redundant roles in development, tissue homeostasis, wound healing, pathology and tumorigenesis of orofacial and dental tissues.
Collapse
Affiliation(s)
- Georgia Nikoloudaki
- Schulich Dentistry Department, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; ; Tel.: +1-519-661-2111 (ext. 81102)
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| |
Collapse
|
44
|
Ustriyana P, Schulte F, Gombedza F, Gil-Bona A, Paruchuri S, Bidlack FB, Hardt M, Landis WJ, Sahai N. Spatial survey of non-collagenous proteins in mineralizing and non-mineralizing vertebrate tissues ex vivo. Bone Rep 2021; 14:100754. [PMID: 33665237 PMCID: PMC7900015 DOI: 10.1016/j.bonr.2021.100754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 11/24/2022] Open
Abstract
Bone biomineralization is a complex process in which type I collagen and associated non-collagenous proteins (NCPs), including glycoproteins and proteoglycans, interact closely with inorganic calcium and phosphate ions to control the precipitation of nanosized, non-stoichiometric hydroxyapatite (HAP, idealized stoichiometry Ca10(PO4)6(OH)2) within the organic matrix of a tissue. The ability of certain vertebrate tissues to mineralize is critically related to several aspects of their function. The goal of this study was to identify specific NCPs in mineralizing and non-mineralizing tissues of two animal models, rat and turkey, and to determine whether some NCPs are unique to each type of tissue. The tissues investigated were rat femur (mineralizing) and tail tendon (non-mineralizing) and turkey leg tendon (having both mineralizing and non-mineralizing regions in the same individual specimen). An experimental approach ex vivo was designed for this investigation by combining sequential protein extraction with comprehensive protein mapping using proteomics and Western blotting. The extraction method enabled separation of various NCPs based on their association with either the extracellular organic collagenous matrix phases or the inorganic mineral phases of the tissues. The proteomics work generated a complete picture of NCPs in different tissues and animal species. Subsequently, Western blotting provided validation for some of the proteomics findings. The survey then yielded generalized results relevant to various protein families, rather than only individual NCPs. This study focused primarily on the NCPs belonging to the small leucine-rich proteoglycan (SLRP) family and the small integrin-binding ligand N-linked glycoproteins (SIBLINGs). SLRPs were found to be associated only with the collagenous matrix, a result suggesting that they are mainly involved in structural matrix organization and not in mineralization. SIBLINGs as well as matrix Gla (γ-carboxyglutamate) protein were strictly localized within the inorganic mineral phase of mineralizing tissues, a finding suggesting that their roles are limited to mineralization. The results from this study indicated that osteocalcin was closely involved in mineralization but did not preclude possible additional roles as a hormone. This report provides for the first time a spatial survey and comparison of NCPs from mineralizing and non-mineralizing tissues ex vivo and defines the proteome of turkey leg tendons as a model for vertebrate mineralization.
Collapse
Key Words
- B, rat bone
- BSP, bone sialoprotein
- DCN, decorin
- E, EDTA extract
- ECM, extracellular matrix
- G, guanidine-HCl-only extract (for non-mineralizing tissues)
- G1, first guanidine-HCl extract
- G2, second guanidine-HCl extract
- Gla, gamma-carboxylated glutamic acid
- MGP, matrix Gla protein
- MT, turkey mineralizing tendon
- Mineralization
- NCP, non-collagenous protein
- NMT, turkey never-mineralizing tendon
- NT, turkey not-yet-mineralized tendon
- Non-collagenous protein
- OCN, osteocalcin
- OPN, osteopontin
- Proteomics
- SIBLING, small integrin-binding ligand N-linked glycoprotein
- SLRP, small leucine-rich proteoglycan
- T, rat tail tendon
- TLT, turkey leg tendon (gastrocnemius)
- TNAP, tissue-nonspecific alkaline phosphatase
- Type I collagen
- Vertebrate
Collapse
Affiliation(s)
- Putu Ustriyana
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH 44325, USA
| | - Fabian Schulte
- The Forsyth Institute, Cambridge, MA 02142, USA
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Farai Gombedza
- Department of Chemistry, The University of Akron, Akron, OH 44325, USA
| | - Ana Gil-Bona
- The Forsyth Institute, Cambridge, MA 02142, USA
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sailaja Paruchuri
- Department of Chemistry, The University of Akron, Akron, OH 44325, USA
| | - Felicitas B. Bidlack
- The Forsyth Institute, Cambridge, MA 02142, USA
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Markus Hardt
- The Forsyth Institute, Cambridge, MA 02142, USA
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - William J. Landis
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH 44325, USA
| | - Nita Sahai
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH 44325, USA
- Department of Geosciences, The University of Akron, Akron, OH 44325, USA
- Integrated Bioscience Program, The University of Akron, Akron, OH 44325, USA
| |
Collapse
|
45
|
Bover J, Ureña-Torres P, Cozzolino M, Rodríguez-García M, Gómez-Alonso C. The Non-invasive Diagnosis of Bone Disorders in CKD. Calcif Tissue Int 2021; 108:512-527. [PMID: 33398414 DOI: 10.1007/s00223-020-00781-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022]
Abstract
Abnormal bone metabolism is an integral part of the chronic kidney disease-mineral bone disorder (CKD-MBD). For several reasons, the difficult bone compartment was neglected for some time, but there has been renewed interest as a result of the conception of bone as a new endocrine organ, the increasing recognition of the cross-talk between bone and vessels, and, especially, the very high risk of osteoporotic fractures (and associated mortality) demonstrated in patients with CKD. Therefore, it has been acknowledged in different guidelines that action is needed in respect of fracture risk assessment and the diagnosis and treatment of osteoporosis in the context of CKD and CKD-MBD, even beyond renal osteodystrophy. These updated guidelines clearly underline the need to improve a non-invasive approach to these bone disorders in order to guide treatment decisions aimed at not only controlling CKD-MBD but also decreasing the risk of fracture. In this report, we review the current role of the most often clinically used or promising biochemical circulating biomarkers such as parathyroid hormone, alkaline phosphatases, and other biochemical markers of bone activity as alternatives to some aspects of bone histomorphometry. We also mention the potential role of classic and new imaging techniques for CKD patients. Information on many aspects is still scarce and heterogeneous, but many of us consider that it is indeed time for action, recognizing our definitely limited ability to base certain treatment decisions only on our current non-comprehensive knowledge.
Collapse
Affiliation(s)
- Jordi Bover
- Department of Nephrology, Fundació Puigvert and Universitat Autònoma, IIB Sant Pau, REDinREN, C. Cartagena 340-350, 08025, Barcelona, Catalonia, Spain.
| | - Pablo Ureña-Torres
- Department of Dialysis, AURA Nord Saint Ouen and Department of Renal Physiology, Necker Hospital, University of Paris Descartes, Paris, France
| | - Mario Cozzolino
- Renal Division, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Minerva Rodríguez-García
- Unidad de Gestión Clínica de Nefrología, Hospital Universitario Central de Asturias, REDinREN, Universidad de Oviedo, Oviedo, Spain
| | - Carlos Gómez-Alonso
- Unidad de Gestión Clínica de Metabolismo Óseo y Mineral, Instituto de Investigación Sanitaria del Principado de Asturias, Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
46
|
Carvalho MS, Cabral JMS, da Silva CL, Vashishth D. Bone Matrix Non-Collagenous Proteins in Tissue Engineering: Creating New Bone by Mimicking the Extracellular Matrix. Polymers (Basel) 2021; 13:polym13071095. [PMID: 33808184 PMCID: PMC8036283 DOI: 10.3390/polym13071095] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023] Open
Abstract
Engineering biomaterials that mimic the extracellular matrix (ECM) of bone is of significant importance since most of the outstanding properties of the bone are due to matrix constitution. Bone ECM is composed of a mineral part comprising hydroxyapatite and of an organic part of primarily collagen with the rest consisting on non-collagenous proteins. Collagen has already been described as critical for bone tissue regeneration; however, little is known about the potential effect of non-collagenous proteins on osteogenic differentiation, even though these proteins were identified some decades ago. Aiming to engineer new bone tissue, peptide-incorporated biomimetic materials have been developed, presenting improved biomaterial performance. These promising results led to ongoing research focused on incorporating non-collagenous proteins from bone matrix to enhance the properties of the scaffolds namely in what concerns cell migration, proliferation, and differentiation, with the ultimate goal of designing novel strategies that mimic the native bone ECM for bone tissue engineering applications. Overall, this review will provide an overview of the several non-collagenous proteins present in bone ECM, their functionality and their recent applications in the bone tissue (including dental) engineering field.
Collapse
Affiliation(s)
- Marta S. Carvalho
- Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (J.M.S.C.); (C.L.d.S.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Correspondence: (M.S.C.); (D.V.)
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (J.M.S.C.); (C.L.d.S.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Cláudia L. da Silva
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (J.M.S.C.); (C.L.d.S.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Deepak Vashishth
- Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Correspondence: (M.S.C.); (D.V.)
| |
Collapse
|
47
|
Hara ES, Okada M, Nagaoka N, Nakano T, Matsumoto T. Re-Evaluation of Initial Bone Mineralization from an Engineering Perspective. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:246-255. [PMID: 33573463 PMCID: PMC8892978 DOI: 10.1089/ten.teb.2020.0352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bone regeneration was one of the earliest fields to develop in the context of tissue regeneration, and currently, repair of small-sized bone defects has reached a high success rate. Future researches are expected to incorporate more advanced techniques toward achieving rapid bone repair and modulation of the regenerated bone quality. For these purposes, it is important to have a more integrative understanding of the mechanisms of bone formation and maturation from multiple perspectives and to incorporate these new concepts into the development and designing of novel materials and techniques for bone regeneration. This review focuses on the analysis of the earliest stages of bone tissue development from the biology, material science, and engineering perspectives for a more integrative understanding of bone formation and maturation, and for the development of novel biology-based engineering approaches for tissue synthesis in vitro. More specifically, the authors describe the systematic methodology that allowed the understanding of the different nucleation sites in intramembranous and endochondral ossification, the space-making process for mineral formation and growth, as well as the process of apatite crystal cluster growth in vivo in the presence of suppressing biomolecules.
Collapse
Affiliation(s)
- Emilio Satoshi Hara
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Okada
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Noriyuki Nagaoka
- Advanced Research Center for Oral & Craniofacial Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takayoshi Nakano
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
48
|
Jablonská E, Horkavcová D, Rohanová D, Brauer DS. A review of in vitro cell culture testing methods for bioactive glasses and other biomaterials for hard tissue regeneration. J Mater Chem B 2021; 8:10941-10953. [PMID: 33169773 DOI: 10.1039/d0tb01493a] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bioactive glasses are used to regenerate bone by a mechanism which involves surface degradation, the release of ions such as calcium, soluble silica and phosphate and the precipitation of a biomimetic apatite surface layer on the glass. One major area of bioactive glass research is the incorporation of therapeutically active ions to broaden the application range of these materials. When developing such new compositions, in vitro cell culture studies are a key part of their characterisation. However, parameters of cell culture studies vary widely, and depending on the intended use of bioactive glass compositions, different layouts, cell types and assays need to be used. The aim of this publication is to provide materials scientists, particularly those new to cell culture studies, with a tool for selecting the most appropriate assays to give insight into the properties of interest.
Collapse
Affiliation(s)
- Eva Jablonská
- Laboratory of Molecular Biology and Virology, Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague 6, Czech Republic.
| | - Diana Horkavcová
- Laboratory of Chemistry and Technology of Glasses, Department of Glass and Ceramics, University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Dana Rohanová
- Laboratory of Chemistry and Technology of Glasses, Department of Glass and Ceramics, University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Delia S Brauer
- Otto Schott Institute of Materials Research, Faculty of Chemistry and Earth Sciences, Friedrich Schiller University Jena, Fraunhoferstr. 6, 07743 Jena, Germany.
| |
Collapse
|
49
|
Kaur S, Roberts DD. Differential intolerance to loss of function and missense mutations in genes that encode human matricellular proteins. J Cell Commun Signal 2021; 15:93-105. [PMID: 33415696 PMCID: PMC7904989 DOI: 10.1007/s12079-020-00598-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
Targeted gene disruption in mice has provided valuable insights into the functions of matricellular proteins. Apart from missense and loss of function mutations that have been associated with inherited diseases, however, their functions in humans remain unclear. The availability of deep exome sequencing data from over 140,000 individuals in the Genome Aggregation Database provided an opportunity to examine intolerance to loss of function and missense mutations in human matricellular genes. The probability of loss-of-function intolerance (pLI) differed widely within members of the thrombospondin, CYR61/CTGF/NOV (CCN), tenascin, small integrin-binding ligand N-linked glycoproteins (SIBLING), and secreted protein, acidic and rich in cysteine (SPARC) gene families. Notably, pLI values in humans had limited correlation with viability of the corresponding homozygous null mice. Among the thrombospondins, only THBS1 was highly loss-intolerant (pLI = 1). In contrast, Thbs1 is not essential for viability in mice. Several known thrombospondin-1 receptors were similarly loss-intolerant, although thrombospondin-1 is not the exclusive ligand for some of these receptors. The frequencies of missense mutations in THBS1 and the gene encoding its signaling receptor CD47 indicated conservation of some residues implicated in specific receptor binding. Deficits in missense mutations were also observed for other thrombospondin genes and for SPARC, SPOCK1, SPOCK2, TNR, and DSPP. The intolerance of THBS1 to loss of function in humans and elevated pLI values for THBS2, SPARC, SPOCK1, TNR, and CCN1 support important functions for these matricellular protein genes in humans, some of which may relate to functions in reproduction or responding to environmental stresses.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Building 10 Room 2S235, 10 Center Drive MSC1500, Bethesda, MD, 20892-1500, USA.
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Building 10 Room 2S235, 10 Center Drive MSC1500, Bethesda, MD, 20892-1500, USA.
| |
Collapse
|
50
|
Sharma A, Goring A, Johnson PB, Emery RJH, Hesse E, Boyde A, Olsen BR, Pitsillides AA, Oreffo ROC, Mahajan S, Clarkin CE. Multiscale molecular profiling of pathological bone resolves sexually dimorphic control of extracellular matrix composition. Dis Model Mech 2021; 14:dmm048116. [PMID: 33563616 PMCID: PMC7988766 DOI: 10.1242/dmm.048116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/21/2021] [Indexed: 11/28/2022] Open
Abstract
Collagen assembly during development is essential for successful matrix mineralisation, which determines bone quality and mechanocompetence. However, the biochemical and structural perturbations that drive pathological skeletal collagen configuration remain unclear. Deletion of vascular endothelial growth factor (VEGF; also known as VEGFA) in bone-forming osteoblasts (OBs) induces sex-specific alterations in extracellular matrix (ECM) conformation and mineralisation coupled to vascular changes, which are augmented in males. Whether this phenotypic dimorphism arises as a result of the divergent control of ECM composition and its subsequent arrangement is unknown and is the focus of this study. Herein, we used murine osteocalcin-specific Vegf knockout (OcnVEGFKO) and performed ex vivo multiscale analysis at the tibiofibular junction of both sexes. Label-free and non-destructive polarisation-resolved second-harmonic generation (p-SHG) microscopy revealed a reduction in collagen fibre number in males following the loss of VEGF, complemented by observable defects in matrix organisation by backscattered electron scanning electron microscopy. This was accompanied by localised divergence in collagen orientation, determined by p-SHG anisotropy measurements, as a result of OcnVEGFKO. Raman spectroscopy confirmed that the effect on collagen was linked to molecular dimorphic VEGF effects on collagen-specific proline and hydroxyproline, and collagen intra-strand stability, in addition to matrix carbonation and mineralisation. Vegf deletion in male and female murine OB cultures in vitro further highlighted divergence in genes regulating local ECM structure, including Adamts2, Spp1, Mmp9 and Lama1. Our results demonstrate the utility of macromolecular imaging and spectroscopic modalities for the detection of collagen arrangement and ECM composition in pathological bone. Linking the sex-specific genetic regulators to matrix signatures could be important for treatment of dimorphic bone disorders that clinically manifest in pathological nano- and macro-level disorganisation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Aikta Sharma
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Alice Goring
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Peter B. Johnson
- School of Chemistry and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Roger J. H. Emery
- Department of Surgery and Cancer, Faculty of Medicine, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Eric Hesse
- Institute of Molecular Musculoskeletal Research, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Munich 80336, Germany
| | - Alan Boyde
- Dental Physical Sciences, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK
| | - Bjorn R. Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Andrew A. Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Richard O. C. Oreffo
- Centre for Human Development, Stem Cell and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Sumeet Mahajan
- School of Chemistry and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Claire E. Clarkin
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|