1
|
Schwartzman JD, McCall M, Ghattas Y, Pugazhendhi AS, Wei F, Ngo C, Ruiz J, Seal S, Coathup MJ. Multifunctional scaffolds for bone repair following age-related biological decline: Promising prospects for smart biomaterial-driven technologies. Biomaterials 2024; 311:122683. [PMID: 38954959 DOI: 10.1016/j.biomaterials.2024.122683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
The repair of large bone defects due to trauma, disease, and infection can be exceptionally challenging in the elderly. Despite best clinical practice, bone regeneration within contemporary, surgically implanted synthetic scaffolds is often problematic, inconsistent, and insufficient where additional osteobiological support is required to restore bone. Emergent smart multifunctional biomaterials may drive important and dynamic cellular crosstalk that directly targets, signals, stimulates, and promotes an innate bone repair response following age-related biological decline and when in the presence of disease or infection. However, their role remains largely undetermined. By highlighting their mechanism/s and mode/s of action, this review spotlights smart technologies that favorably align in their conceivable ability to directly target and enhance bone repair and thus are highly promising for future discovery for use in the elderly. The four degrees of interactive scaffold smartness are presented, with a focus on bioactive, bioresponsive, and the yet-to-be-developed autonomous scaffold activity. Further, cell- and biomolecular-assisted approaches were excluded, allowing for contemporary examination of the capabilities, demands, vision, and future requisites of next-generation biomaterial-induced technologies only. Data strongly supports that smart scaffolds hold significant promise in the promotion of bone repair in patients with a reduced osteobiological response. Importantly, many techniques have yet to be tested in preclinical models of aging. Thus, greater clarity on their proficiency to counteract the many unresolved challenges within the scope of aging bone is highly warranted and is arguably the next frontier in the field. This review demonstrates that the use of multifunctional smart synthetic scaffolds with an engineered strategy to circumvent the biological insufficiencies associated with aging bone is a viable route for achieving next-generation therapeutic success in the elderly population.
Collapse
Affiliation(s)
| | - Max McCall
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Yasmine Ghattas
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Abinaya Sindu Pugazhendhi
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Fei Wei
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Christopher Ngo
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Jonathan Ruiz
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA; Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, USA, Orlando, FL
| | - Melanie J Coathup
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
2
|
Zhu J, Du W, Zeng Y, Liu T, Li J, Wang A, Li Y, Zhang W, Huang JA, Liu Z. CD73 promotes non-small cell lung cancer metastasis by regulating Axl signaling independent of GAS6. Proc Natl Acad Sci U S A 2024; 121:e2404709121. [PMID: 39423241 PMCID: PMC11513981 DOI: 10.1073/pnas.2404709121] [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: 03/06/2024] [Accepted: 09/12/2024] [Indexed: 10/21/2024] Open
Abstract
As catabolic enzyme, CD73 dephosphorylates adenosine monophosphate (AMP) and can also regulate tumor cell proliferation and metastasis. To date, very few studies have explored the role of CD73 in mediating non-small cell lung cancer (NSCLC) metastasis, and the underlying transducing signal has not been elucidated. In the present study, we demonstrated that the CD73/Axl axis could regulate Smad3-induced epithelial-to-mesenchymal transition (EMT) to promote NSCLC metastasis. Mechanically, CD73 can be secreted via the Golgi apparatus transport pathway. Then secreted CD73 may activate AXl by directly bind with site R55 located in Axl extracellular domain independently of GAS6. In addition, we proved that CD73 can stabilize Axl expression via inhibiting CBLB expression. We also identified the distinct function of CD73 activity in adenocarcinoma and squamous cell carcinoma. Our findings indicated a role of CD73 in mediating NSCLC metastasis and propose it as a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Jianjie Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Wenwen Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Yuanyuan Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Ting Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
| | - Jianjun Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Anqi Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Yue Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
| | - Weijie Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
| | - Jian-an Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| |
Collapse
|
3
|
Beeraka NM, Basappa B, Nikolenko VN, Mahesh PA. Role of Neurotransmitters in Steady State Hematopoiesis, Aging, and Leukemia. Stem Cell Rev Rep 2024:10.1007/s12015-024-10761-z. [PMID: 38976142 DOI: 10.1007/s12015-024-10761-z] [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] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Haematopoiesis within the bone marrow (BM) represents a complex and dynamic process intricately regulated by neural signaling pathways. This delicate orchestration is susceptible to disruption by factors such as aging, diabetes, and obesity, which can impair the BM niche and consequently affect haematopoiesis. Genetic mutations in Tet2, Dnmt3a, Asxl1, and Jak2 are known to give rise to clonal haematopoiesis of intermediate potential (CHIP), a condition linked to age-related haematological malignancies. Despite these insights, the exact roles of circadian rhythms, sphingosine-1-phosphate (S1P), stromal cell-derived factor-1 (SDF-1), sterile inflammation, and the complement cascade on various BM niche cells remain inadequately understood. Further research is needed to elucidate how BM niche cells contribute to these malignancies through neural regulation and their potential in the development of gene-corrected stem cells. This literature review describes the updated functional aspects of BM niche cells in haematopoiesis within the context of haematological malignancies, with a particular focus on neural signaling and the potential of radiomitigators in acute radiation syndrome. Additionally, it underscores the pressing need for technological advancements in stem cell-based therapies to alleviate the impacts of immunological stressors. Recent studies have illuminated the microheterogeneity and temporal stochasticity of niche cells within the BM during haematopoiesis, emphasizing the updated roles of neural signaling and immunosurveillance. The development of gene-corrected stem cells capable of producing blood, immune cells, and tissue-resident progeny is essential for combating age-related haematological malignancies and overcoming immunological challenges. This review aims to provide a comprehensive overview of these evolving insights and their implications for future therapeutic strategies.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN, 46202, USA.
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia.
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India.
| | - Basappa Basappa
- Department of Studies in Organic Chemistry, Laboratory of Chemical Biology, University of Mysore, Mysore, Karnataka, 570006, India
| | - Vladimir N Nikolenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia
| | - P A Mahesh
- Department of Pulmonary Medicine, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| |
Collapse
|
4
|
Ehlen QT, Mirsky NA, Slavin BV, Parra M, Nayak VV, Cronstein B, Witek L, Coelho PG. Translational Experimental Basis of Indirect Adenosine Receptor Agonist Stimulation for Bone Regeneration: A Review. Int J Mol Sci 2024; 25:6104. [PMID: 38892291 PMCID: PMC11172580 DOI: 10.3390/ijms25116104] [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: 04/10/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Bone regeneration remains a significant clinical challenge, often necessitating surgical approaches when healing bone defects and fracture nonunions. Within this context, the modulation of adenosine signaling pathways has emerged as a promising therapeutic option, encouraging osteoblast activation and tempering osteoclast differentiation. A literature review of the PubMed database with relevant keywords was conducted. The search criteria involved in vitro or in vivo models, with clear methodological descriptions. Only studies that included the use of indirect adenosine agonists, looking at the effects of bone regeneration, were considered relevant according to the eligibility criteria. A total of 29 articles were identified which met the inclusion and exclusion criteria, and they were reviewed to highlight the preclinical translation of adenosine agonists. While preclinical studies demonstrate the therapeutic potential of adenosine signaling in bone regeneration, its clinical application remains unrealized, underscoring the need for further clinical trials. To date, only large, preclinical animal models using indirect adenosine agonists have been successful in stimulating bone regeneration. The adenosine receptors (A1, A2A, A2B, and A3) stimulate various pathways, inducing different cellular responses. Specifically, indirect adenosine agonists act to increase the extracellular concentration of adenosine, subsequently agonizing the respective adenosine receptors. The agonism of each receptor is dependent on its expression on the cell surface, the extracellular concentration of adenosine, and its affinity for adenosine. This comprehensive review analyzed the multitude of indirect agonists currently being studied preclinically for bone regeneration, discussing the mechanisms of each agonist, their cellular responses in vitro, and their effects on bone formation in vivo.
Collapse
Affiliation(s)
- Quinn T. Ehlen
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Blaire V. Slavin
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Marcelo Parra
- Center of Excellence in Morphological and Surgical Studies (CEMyQ), Faculty of Medicine, Universidad de la Frontera, Temuco 4811230, Chile
- Department of Comprehensive Adult Dentistry, Faculty of Dentistry, Universidad de la Frontera, Temuco 4811230, Chile
| | - Vasudev Vivekanand Nayak
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Bruce Cronstein
- Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Lukasz Witek
- Biomaterials Division, NYU Dentistry, New York, NY 10010, USA
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Paulo G. Coelho
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
5
|
Li GHY, Tang CM, Wu SM, Cheung CL. Causal association of genetically determined caffeine intake from tea or coffee with bone health: a two-sample Mendelian randomization study. Postgrad Med J 2024:qgae051. [PMID: 38651568 DOI: 10.1093/postmj/qgae051] [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: 10/17/2023] [Revised: 02/24/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Relationship of caffeine intake and consumption of caffeinated beverages, such as tea and coffee, with bone health remains controversial. This study aimed to evaluate whether genetically determined caffeine intake from tea or coffee has causal effects on overall total body bone mineral density (TB-BMD) and fracture. We also assessed the association with TB-BMD in five age strata. METHODS Using two-sample Mendelian randomization approach, summary statistics were retrieved from genome-wide association studies (GWAS)/GWAS meta-analyses of caffeine intake from tea (n = 395 866)/coffee (n = 373 522), TB-BMD (n = 66 628), and fracture (n = 426 795). Inverse variance weighted method was adopted as the main univariable analysis. Multivariable analysis was conducted to evaluate whether the causal effect is independent. RESULTS In univariable analysis, genetically determined caffeine intake from tea had positive association with overall TB-BMD (per SD increase in genetically determined caffeine intake, beta of TB-BMD [in SD]: 0.166; 95% confidence interval (CI): 0.006-0.326) and inverse association with fracture (OR = 0.79; 95% CI: 0.654-0.954). Genetically determined caffeine intake from coffee was also positively associated with overall TB-BMD (beta = 0.231; 95% CI: 0.093-0.369). The association remained significant after adjustment for smoking in multivariable analysis. Genetically determined caffeine intake from tea or coffee was both positively associated with TB-BMD in the age strata of 45-60 years, but we lacked evidence of association in other strata. CONCLUSIONS Genetically, caffeine intake from tea or coffee may be beneficial to bone health. Due to the ascertainment method of caffeine intake from tea, our study also implied genetically higher tea consumption may improve TB-BMD and lower fracture risk.
Collapse
Affiliation(s)
- Gloria Hoi-Yee Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Lee Shau Kee Building, 11 Yuk Choi Road, Hung Hom, Hong Kong
| | - Ching-Man Tang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Lee Shau Kee Building, 11 Yuk Choi Road, Hung Hom, Hong Kong
| | - Suet-Man Wu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Lee Shau Kee Building, 11 Yuk Choi Road, Hung Hom, Hong Kong
| | - Ching-Lung Cheung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong
| |
Collapse
|
6
|
Ramos-Junior ES, Dawson S, Ryan W, Clinebell B, Serrano-Lopez R, Russell M, Brumbaugh R, Zhong R, Gonçalves Fernandes J, Shaddox LM, Cutler CW, Morandini AC. The protective role of CD73 in periodontitis: preventing hyper-inflammatory fibroblasts and driving osteoclast energy metabolism. FRONTIERS IN ORAL HEALTH 2023; 4:1308657. [PMID: 38152410 PMCID: PMC10751373 DOI: 10.3389/froh.2023.1308657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/21/2023] [Indexed: 12/29/2023] Open
Abstract
Introduction Periodontitis is an immune-mediated inflammatory disease affecting almost half of the adult population and is the leading cause of tooth loss in the United States. The role of extracellular nucleotide signaling including nucleotide metabolizing enzyme CD73 adds an important layer of interaction of purine mediators capable of orchestrating inflammatory outcomes. CD73 is able to catabolize 5'-adenosine monophosphate into adenosine at the extracellular level, playing a critical role in regulating many processes under physiological and pathological conditions. Here, we explored the role of CD73 in ligature-induced periodontitis in vivo comparing wild-type C57Bl/6J and CD73-deficient mice. Methods We assessed gingival levels of inflammatory cytokines in vivo and in murine gingival fibroblasts in vitro, as well as bone loss, and RANKL-induced osteoclastogenesis. We have also analyzed CD73 mRNA in samples derived from patients diagnosed with severe periodontitis. Results Our results in mice show that lack of CD73 resulted in increased inflammatory cytokines and chemokines such as IL-1β, IL-17, Cxcl1 and Cxcl2 in diseased gingiva relative to the healthy-controls and in comparison with the wild type. CD73-deficient gingival fibroblasts also manifested a defective healing response with higher MMP-13 levels. CD73-deficient animals also showed increased osteoclastogenesis in vitro with increased mitochondrial metabolism typified by excessive activation of oxidative phosphorylation, increased mitochondrial membrane potential and accumulation of hydrogen peroxide. Micro-CT analysis revealed that lack of CD73 resulted in decreased bone mineral density, decreased trabecular bone volume and thickness as well as decreased bone volume in long bones. CD73 deficiency also resulted in increased alveolar bone loss in experimental periodontitis. Correlative studies of gingival samples from severe (Grade C) periodontitis showed decreased levels of CD73 compared to healthy controls, further supporting the relevance of our murine results. Conclusion In conclusion, CD73 appears to play a protective role in the gingival periodontal tissue and bone homeostasis, regulating hyper-inflammatory state of stromal fibroblasts and osteoclast energy metabolism and being an important candidate for future target therapies to prevent or control immune-mediated inflammatory and osteolytic diseases.
Collapse
Affiliation(s)
- Erivan S. Ramos-Junior
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Shantiece Dawson
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Weston Ryan
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Braden Clinebell
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Rogelio Serrano-Lopez
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Marsha Russell
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Rylee Brumbaugh
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Roger Zhong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jussara Gonçalves Fernandes
- Division of Periodontology and Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Luciana M. Shaddox
- Division of Periodontology and Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Christopher W. Cutler
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Ana Carolina Morandini
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
7
|
Najjary S, de Koning W, Kros JM, Mustafa DAM. Unlocking molecular mechanisms and identifying druggable targets in matched-paired brain metastasis of breast and lung cancers. Front Immunol 2023; 14:1305644. [PMID: 38149244 PMCID: PMC10750385 DOI: 10.3389/fimmu.2023.1305644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/16/2023] [Indexed: 12/28/2023] Open
Abstract
Introduction The incidence of brain metastases in cancer patients is increasing, with lung and breast cancer being the most common sources. Despite advancements in targeted therapies, the prognosis remains poor, highlighting the importance to investigate the underlying mechanisms in brain metastases. The aim of this study was to investigate the differences in the molecular mechanisms involved in brain metastasis of breast and lung cancers. In addition, we aimed to identify cancer lineage-specific druggable targets in the brain metastasis. Methods To that aim, a cohort of 44 FFPE tissue samples, including 22 breast cancer and 22 lung adenocarcinoma (LUAD) and their matched-paired brain metastases were collected. Targeted gene expression profiles of primary tumors were compared to their matched-paired brain metastases samples using nCounter PanCancer IO 360™ Panel of NanoString technologies. Pathway analysis was performed using gene set analysis (GSA) and gene set enrichment analysis (GSEA). The validation was performed by using Immunohistochemistry (IHC) to confirm the expression of immune checkpoint inhibitors. Results Our results revealed the significant upregulation of cancer-related genes in primary tumors compared to their matched-paired brain metastases (adj. p ≤ 0.05). We found that upregulated differentially expressed genes in breast cancer brain metastasis (BM-BC) and brain metastasis from lung adenocarcinoma (BM-LUAD) were associated with the metabolic stress pathway, particularly related to the glycolysis. Additionally, we found that the upregulated genes in BM-BC and BM-LUAD played roles in immune response regulation, tumor growth, and proliferation. Importantly, we identified high expression of the immune checkpoint VTCN1 in BM-BC, and VISTA, IDO1, NT5E, and HDAC3 in BM-LUAD. Validation using immunohistochemistry further supported these findings. Conclusion In conclusion, the findings highlight the significance of using matched-paired samples to identify cancer lineage-specific therapies that may improve brain metastasis patients outcomes.
Collapse
Affiliation(s)
| | | | | | - Dana A. M. Mustafa
- Department of Pathology and Clinical Bioinformatics, The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
8
|
Pinto-Cardoso R, Bessa-Andrês C, Correia-de-Sá P, Bernardo Noronha-Matos J. Could hypoxia rehabilitate the osteochondral diseased interface? Lessons from the interplay of hypoxia and purinergic signals elsewhere. Biochem Pharmacol 2023:115646. [PMID: 37321413 DOI: 10.1016/j.bcp.2023.115646] [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: 04/07/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
The osteochondral unit comprises the articular cartilage (90%), subchondral bone (5%) and calcified cartilage (5%). All cells present at the osteochondral unit that is ultimately responsible for matrix production and osteochondral homeostasis, such as chondrocytes, osteoblasts, osteoclasts and osteocytes, can release adenine and/or uracil nucleotides to the local microenvironment. Nucleotides are released by these cells either constitutively or upon plasma membrane damage, mechanical stress or hypoxia conditions. Once in the extracellular space, endogenously released nucleotides can activate membrane-bound purinoceptors. Activation of these receptors is fine-tuning regulated by nucleotides' breakdown by enzymes of the ecto-nucleotidase cascade. Depending on the pathophysiological conditions, both the avascular cartilage and the subchondral bone subsist to significant changes in oxygen tension, which has a tremendous impact on tissue homeostasis. Cell stress due to hypoxic conditions directly influences the expression and activity of several purinergic signalling players, namely nucleotide release channels (e.g. Cx43), NTPDase enzymes and purinoceptors. This review gathers experimental evidence concerning the interplay between hypoxia and the purinergic signalling cascade contributing to osteochondral unit homeostasis. Reporting deviations to this relationship resulting from pathological alterations of articular joints may ultimately unravel novel therapeutic targets for osteochondral rehabilitation. At this point, one can only hypothesize how hypoxia mimetic conditions can be beneficial to the ex vivo expansion and differentiation of osteo- and chondro-progenitors for auto-transplantation and tissue regenerative purposes.
Collapse
Affiliation(s)
- Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP).
| |
Collapse
|
9
|
Newman H, Varghese S. Extracellular adenosine signaling in bone health and disease. Curr Opin Pharmacol 2023; 70:102378. [PMID: 37044008 PMCID: PMC10247430 DOI: 10.1016/j.coph.2023.102378] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/29/2023] [Accepted: 03/13/2023] [Indexed: 04/14/2023]
Abstract
Purinergic signaling is a key molecular pathway in the maintenance of bone health and regeneration. P1 receptor signaling, which is activated by extracellular adenosine, has emerged as a key metabolic pathway that regulates bone tissue formation, function, and homeostasis. Extracellular adenosine is mainly produced by ectonucleotidases, and alterations in the function of these enzymes or compromised adenosine generation can result in bone disorders, such as osteoporosis and impaired fracture healing. This mini review discusses the key role played by adenosine in bone health and how its alterations contribute to bone diseases, as well as potential therapeutic applications of exogenous adenosine to combat bone diseases like osteoporosis and injury.
Collapse
Affiliation(s)
- Hunter Newman
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA; Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shyni Varghese
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA; Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
10
|
Larrañaga-Vera A, Toti KS, Flatow JS, Haraczy AJ, Warnick E, Rao H, Gao ZG, Sussman SM, Mediero A, Leucht P, Jacobson KA, Cronstein BN. Novel alendronate-CGS21680 conjugate reduces bone resorption and induces new bone formation in post-menopausal osteoporosis and inflammatory osteolysis mouse models. Arthritis Res Ther 2022; 24:265. [PMID: 36494860 PMCID: PMC9733060 DOI: 10.1186/s13075-022-02961-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Loss of bone is a common medical problem and, while it can be treated with available therapies, some of these therapies have critical side effects. We have previously demonstrated that CGS21680, a selective A2A adenosine receptor agonist, prevents bone loss, but its on-target toxicities (hypotension, tachycardia) and frequent dosing requirements make it unusable in the clinic. We therefore generated a novel alendronate-CGS21680 conjugate (MRS7216), to target the agonist to bone where it remains for long periods thereby diminishing the frequency of administration and curtailing side effects. MRS7216 was synthesized from CGS21680 by sequential activation of the carboxylic acid moiety and reacting with an appropriate amino acid (PEG, alendronic acid) under basic conditions. MRS7216 was tested on C57BL/6J (WT) mice with established osteoporosis (OP) and WT or A2A KO mice with wear particle-induced inflammatory osteolysis (OL). Mice were treated weekly with MRS7216 (10mg/kg). Bone formation was studied after in vivo labeling with calcein/Alizarin Red, and μCT and histology analyses were performed. In addition, human primary osteoblasts and osteoclasts were cultured using bone marrow discarded after hip replacement. Receptor binding studies demonstrate that MRS7216 efficiently binds the A2A adenosine receptor. MRS7216-treated OP and OL mice had significant new bone formation and reduced bone loss compared to vehicle or alendronate-treated mice. Histological analysis showed that MRS7216 treatment significantly reduced osteoclast number and increased osteoblast number in murine models. Interestingly, cultured human osteoclast differentiation was inhibited, and osteoblast differentiation was stimulated by the compound indicating that MRS7216 conjugates represent a novel therapeutic approach to treat osteoporosis and osteolysis.
Collapse
Affiliation(s)
- Ane Larrañaga-Vera
- Division of Translational Medicine, Department of Medicine, NYU Langone Health, 550 First Avenue, Medical Science Building, Room, New York, NY, 251, USA
| | - Kiran S Toti
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - James S Flatow
- Division of Translational Medicine, Department of Medicine, NYU Langone Health, 550 First Avenue, Medical Science Building, Room, New York, NY, 251, USA
| | - Alexandra J Haraczy
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Eugene Warnick
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Harsha Rao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah M Sussman
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Aranzazu Mediero
- Division of Translational Medicine, Department of Medicine, NYU Langone Health, 550 First Avenue, Medical Science Building, Room, New York, NY, 251, USA
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Philipp Leucht
- Department of Orthopedic Surgery, New York University Langone Orthopedic Hospital, New York, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bruce N Cronstein
- Division of Translational Medicine, Department of Medicine, NYU Langone Health, 550 First Avenue, Medical Science Building, Room, New York, NY, 251, USA.
- Division of Rheumatology, Department of Medicine, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
11
|
The Inhibition of the Inducible Nitric Oxide Synthase Enhances the DPSC Mineralization under LPS-Induced Inflammation. Int J Mol Sci 2022; 23:ijms232314560. [PMID: 36498888 PMCID: PMC9736592 DOI: 10.3390/ijms232314560] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
Nitric oxide (NO) is a key messenger in physiological and pathological processes in mammals. An excessive NO production is associated with pathological conditions underlying the inflammation response as a trigger. Among others, dental pulp inflammation results from the invasion of dentin by pathogenic bacteria. Vital functions of pulp mesenchymal stem cells (DPSCs, dental pulp stem cells), such as mineralization, might be affected by the inducible NOS (iNOS) upregulation. In this context, the iNOS selective inhibition can be considered an innovative therapeutic strategy to counteract inflammation and to promote the regeneration of the dentin-pulp complex. The present work aims at evaluating two acetamidines structurally related to the selective iNOS inhibitor 1400W, namely CM544 and FAB1020, in a model of LPS-stimulated primary DPSCs. Our data reveal that CM544 and even more FAB1020 are promising anti-inflammatory compounds, decreasing IL-6 secretion by enhancing CD73 expression-levels, a protein involved in innate immunity processes and thus confirming an immunomodulatory role of DPSCs. In parallel, cell mineralization potential is retained in the presence of compounds as well as VEGF secretion, and thus their angiogenetic potential. Data presented lay the ground for further investigation on the anti-inflammatory potential of acetamidines selectively targeting iNOS in a clinical context.
Collapse
|
12
|
Hoque J, Zeng Y, Newman H, Gonzales G, Lee C, Varghese S. Microgel-Assisted Delivery of Adenosine to Accelerate Fracture Healing. ACS Biomater Sci Eng 2022; 8:4863-4872. [PMID: 36266245 PMCID: PMC11188841 DOI: 10.1021/acsbiomaterials.2c00977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Extracellular adenosine plays a key role in promoting bone tissue formation. Local delivery of adenosine could be an effective therapeutic strategy to harness the beneficial effect of extracellular adenosine on bone tissue formation following injury. Herein, we describe the development of an injectable in situ curing scaffold containing microgel-based adenosine delivery units. The two-component scaffold includes adenosine-loaded microgels and functionalized hyaluronic acid (HA) molecules. The microgels were generated upon copolymerization of 3-acrylamidophenylboronic acid (3-APBA)- and 2-aminoethylmethacrylamide (2-AEMA)-conjugated HA (HA-AEMA) in an emulsion suspension. The PBA functional groups were used to load the adenosine molecules. Mixing of the microgels with the HA polymers containing clickable groups, dibenzocyclooctyne (DBCO) and azide (HA-DBCO and HA-Azide), resulted in a 3D scaffold embedded with adenosine delivery units. Application of the in situ curing scaffolds containing adenosine-loaded microgels following tibial fracture injury showed improved bone tissue healing in a mouse model as demonstrated by the reduced callus size, higher bone volume, and increased tissue mineral density compared to those treated with the scaffold without adenosine. Overall, our results suggest that local delivery of adenosine could potentially be an effective strategy to promote bone tissue repair.
Collapse
Affiliation(s)
- Jiaul Hoque
- Department of Orthopaedic Surgery School of Medicine, Duke University, Durham, North Carolina 27710, United States
| | - Yuze Zeng
- Department of Orthopaedic Surgery School of Medicine, Duke University, Durham, North Carolina 27710, United States
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27710, United States
| | - Hunter Newman
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27710, United States
| | - Gavin Gonzales
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27710, United States
| | - Cheryl Lee
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27710, United States
| | - Shyni Varghese
- Department of Orthopaedic Surgery School of Medicine, Duke University, Durham, North Carolina 27710, United States
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27710, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27710, United States
| |
Collapse
|
13
|
Simões AP, Gonçalves FQ, Rial D, Ferreira SG, Lopes JP, Canas PM, Cunha RA. CD73-Mediated Formation of Extracellular Adenosine Is Responsible for Adenosine A 2A Receptor-Mediated Control of Fear Memory and Amygdala Plasticity. Int J Mol Sci 2022; 23:12826. [PMID: 36361618 PMCID: PMC9653840 DOI: 10.3390/ijms232112826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 08/27/2023] Open
Abstract
Adenosine A2A receptors (A2AR) control fear memory and the underlying processes of synaptic plasticity in the amygdala. In other brain regions, A2AR activation is ensured by ATP-derived extracellular adenosine formed by ecto-5'-nucleotidase or CD73. We now tested whether CD73 is also responsible to provide for the activation of A2AR in controlling fear memory and amygdala long-term potentiation (LTP). The bilateral intracerebroventricular injection of the CD73 inhibitor αβ-methylene ADP (AOPCP, 1 nmol/ventricle/day) phenocopied the effect of the A2AR blockade by decreasing the expression of fear memory, an effect disappearing in CD73-knockout (KO) mice and in forebrain neuronal A2AR-KO mice. In the presence of PPADS (20 μM) to eliminate any modification of ATP/ADP-mediated P2 receptor effects, both AOPCP (100 μM) and the A2AR antagonist, SCH58261 (50 nM), decreased LTP magnitude in synapses of projection from the external capsula into the lateral amygdala, an effect eliminated in slices from both forebrain neuronal A2AR-KO mice and CD73-KO mice. These data indicate a key role of CD73 in the process of A2AR-mediated control of fear memory and underlying synaptic plasticity processes in the amygdala, paving the way to envisage CD73 as a new therapeutic target to interfere with abnormal fear-like emotional processing.
Collapse
Affiliation(s)
- Ana Patrícia Simões
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Francisco Q. Gonçalves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Daniel Rial
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Samira G. Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - João Pedro Lopes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paula M. Canas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Rodrigo A. Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
14
|
Newman H, Hoque J, Shih YRV, Marushack G, Ko U, Gonzales G, Varghese S. pH-Sensitive nanocarrier assisted delivery of adenosine to treat osteoporotic bone loss. Biomater Sci 2022; 10:5340-5355. [PMID: 35929516 PMCID: PMC9894097 DOI: 10.1039/d2bm00843b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Bone tissue undergoes continuous remodeling via osteoclast-mediated bone resorption and osteoblast-mediated bone formation. An imbalance in this process with enhanced osteoclastic activity can lead to excessive bone resorption, resulting in bone thinning. Once activated, osteoclasts bind to the bone surface and acidify the local niche. This acidic environment could serve as a potential trigger for the delivery of therapeutic agents into the osteoporotic bone tissue. To this end, we developed a pH-responsive nanocarrier-based drug delivery system that binds to the bone tissue and delivers an osteoanabolic molecule, adenosine. Adenosine is incorporated into a hyaluronic acid (HA)-based nanocarrier through a pH-sensitive ketal group. The HA-nanocarrier is further functionalized with alendronate moieties to improve binding to the bone tissues. Systemic administration of the nanocarrier containing adenosine attenuated bone loss in ovariectomized mice and showed comparable bone qualities to that of healthy mice. Delivery of osteoanabolic small molecules that can contribute to bone formation and inhibit excessive osteoclast activity by leveraging the tissue-specific milieu could serve as viable therapeutics for osteoporosis.
Collapse
Affiliation(s)
- Hunter Newman
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA.
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jiaul Hoque
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yu-Ru V Shih
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gabrielle Marushack
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Unghyeon Ko
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gavin Gonzales
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Shyni Varghese
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA.
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| |
Collapse
|
15
|
Velankar KY, Mou M, Hartmeier PR, Clegg B, Gawalt ES, Jiang M, Meng WS. Recrystallization of Adenosine for Localized Drug Delivery. Mol Pharm 2022; 19:3394-3404. [PMID: 36001090 DOI: 10.1021/acs.molpharmaceut.2c00527] [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: 11/29/2022]
Abstract
Adenosine (ADO) is an endogenous metabolite with immense potential to be repurposed as an immunomodulatory therapeutic, as preclinical studies have demonstrated in models of epilepsy, acute respiratory distress syndrome, and traumatic brain injury, among others. The currently licensed products Adenocard and Adenoscan are formulated at 3 mg/mL of ADO for rapid bolus intravenous injection, but the systemic administration of the saline formulations for anti-inflammatory purposes is limited by the nucleoside's profound hemodynamic effects. Moreover, concentrations that can be attained in the airway or the brain through direct instillation or injection are limited by the volumes that can be accommodated in the anatomical space (<5 mL in humans) and the rapid elimination by enzymatic and transport mechanisms in the interstitium (half-life <5 s). As such, highly concentrated formulations of ADO are needed to attain pharmacologically relevant concentrations at sites of tissue injury. Herein, we report a previously uncharacterized crystalline form of ADO (rcADO) in which 6.7 mg/mL of the nucleoside is suspended in water. Importantly, the crystallinity is not diminished in a protein-rich environment, as evidenced by resuspending the crystals in albumin (15% w/v). To the best of our knowledge, this is the first report of crystalline ADO generated using a facile and organic solvent-free method aimed at localized drug delivery. The crystalline suspension may be suitable for developing ADO into injectable formulations for attaining high concentrations of the endogenous nucleoside in inflammatory locales.
Collapse
Affiliation(s)
- Ketki Y Velankar
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Mingyao Mou
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Paul R Hartmeier
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Benjamin Clegg
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Ellen S Gawalt
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, Pennsylvania 15282, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| | - Mo Jiang
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23284, United States.,Center for Pharmaceutical Engineering and Sciences, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| |
Collapse
|
16
|
Dsouza C, Moussa MS, Mikolajewicz N, Komarova SV. Extracellular ATP and its derivatives provide spatiotemporal guidance for bone adaptation to wide spectrum of physical forces. Bone Rep 2022; 17:101608. [PMID: 35992507 PMCID: PMC9385560 DOI: 10.1016/j.bonr.2022.101608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
ATP is a ubiquitous intracellular molecule critical for cellular bioenergetics. ATP is released in response to mechanical stimulation through vesicular release, small tears in cellular plasma membranes, or when cells are destroyed by traumatic forces. Extracellular ATP is degraded by ecto-ATPases to form ADP and eventually adenosine. ATP, ADP, and adenosine signal through purinergic receptors, including seven P2X ATP-gated cation channels, seven G-protein coupled P2Y receptors responsive to ATP and ADP, and four P1 receptors stimulated by adenosine. The goal of this review is to build a conceptual model of the role of different components of this complex system in coordinating cellular responses that are appropriate to the degree of mechanical stimulation, cell proximity to the location of mechanical injury, and time from the event. We propose that route and amount of ATP release depend on the scale of mechanical forces, ranging from vesicular release of small ATP boluses upon membrane deformation, to leakage of ATP through resealable plasma membrane tears, to spillage of cellular content due to destructive forces. Correspondingly, different P2 receptors responsive to ATP will be activated according to their affinity at the site of mechanical stimulation. ATP is a small molecule that readily diffuses through the environment, bringing the signal to the surrounding cells. ATP is also degraded to ADP which can stimulate a distinct set of P2 receptors. We propose that depending on the magnitude of mechanical forces and distance from the site of their application, ATP/ADP profiles will be different, allowing the relay of information about tissue level injury and proximity. Lastly, ADP is degraded to adenosine acting via its P1 receptors. The presence of large amounts of adenosine without ATP, indicates that an active source of ATP release is no longer present, initiating the transition to the recovery phase. This model consolidates the knowledge regarding the individual components of the purinergic system into a conceptual framework of choreographed responses to physical forces. Cellular bioenergetic molecule ATP is released when cell is mechanically stimulated. ATP release is proportional to the amount of cellular damage. ATP diffusion and transformation to ADP indicates the proximity to the damage. Purinergic receptors form a network choreographing cell response to physical forces. Complete transformation of ATP to adenosine initiates the recovery phase.
Collapse
Affiliation(s)
- Chrisanne Dsouza
- Department of Experimental Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
| | - Mahmoud S. Moussa
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| | - Nicholas Mikolajewicz
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| | - Svetlana V. Komarova
- Department of Experimental Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
- Corresponding author.
| |
Collapse
|
17
|
Zhang H, Cao Y, Tang J, Wang R. CD73 (NT5E) Promotes the Proliferation and Metastasis of Lung Adenocarcinoma through the EGFR/AKT/mTOR Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9944847. [PMID: 35813221 PMCID: PMC9259339 DOI: 10.1155/2022/9944847] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/11/2022] [Indexed: 12/24/2022]
Abstract
Introduction Lung cancer is the most common malignant tumor and the main cause of tumor-related death globally. As the 5-year survival rate of lung adenocarcinoma (LUAD) remains low, it is necessary to investigate novel molecular markers and therapeutic targets for LUAD. Materials and Methods The protein expression of CD73 (NT5E) in LUAD specimens was analyzed using immunohistochemistry. Reverse transcription-quantitative PCR and western blot analysis were used to analyze the mRNA and protein expression levels of several genes in LUAD cells. The proliferation of LUAD cells was evaluated using proliferation and colony formation assays and apoptosis analysis. Wound healing and Transwell invasion assays were used to analyze the migration and invasion of the A549 cells, respectively. In addition, overexpression plasmids and small interfering RNAs were used to overexpress or knockdown the expression levels of CD73 in the A549 cell line, respectively. Finally, the interaction between CD73 and EGFR in the A549 cell line was analyzed using immunoprecipitation. Results Our research emphasized the importance of CD73 in the prognosis of LUAD and highlighted it as a potential therapeutic target. We also found that the mRNA and protein expression levels of CD73 are increased in LUAD specimens and cell lines and were associated with a poor prognosis in patients with LUAD. Furthermore, it was revealed that CD73 may promote the proliferation, migration, and invasion of the A549 cell line. Finally, we demonstrated that CD73 could bind epidermal growth factor receptor (EGFR) to further regulate the activation of the AKT/mTOR signaling pathway. Conclusions CD73 promotes LUAD proliferation and metastasis through EGFR/AKT/mTOR axis.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, No. 1, Shuanghu Branch Road, Yubei District, Chongqing 401120, China
| | - Yu Cao
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, No. 1, Shuanghu Branch Road, Yubei District, Chongqing 401120, China
| | - Jianming Tang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, No. 1, Shuanghu Branch Road, Yubei District, Chongqing 401120, China
| | - Rui Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, No. 1, Shuanghu Branch Road, Yubei District, Chongqing 401120, China
| |
Collapse
|
18
|
CD73/Adenosine Pathway Involvement in the Interaction of Non-Small Cell Lung Cancer Stem Cells and Bone Cells in the Pre-Metastatic Niche. Int J Mol Sci 2022; 23:ijms23095126. [PMID: 35563517 PMCID: PMC9104817 DOI: 10.3390/ijms23095126] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
Adenosinergic signaling is an important regulator of tissue homeostasis and extracellular accumulation of adenosine (Ado) and is associated with different pathologies, such as cancer. In non-small-cell lung cancer (NSCLC), a subset of CD133/CXCR4+ cancer stem cell (CSCs) has been demonstrated to initiate bone metastases. Here we investigated how NSCLC CSCs interact with osteoclasts (OCs) and osteoblasts (OBs) by modulating Ado production and OC activity. We proved that CSC-spheres, generated in vitro from NSCLC cell lines, express CD38, PC-1, and CD73, enzymes of the non-canonical adenosinergic pathway, produce high level of Ado, and down-regulate A1R and A3R inhibitory receptors, while expressing A2AR and A2BR. To address the Ado role and modulation of the in-bone pre-metastatic niche, we performed co-cultures of CSC-spheres with OCs and OBs cells. Firstly, we verified that active OCs do not activate non-canonical the adenosinergic pathway, conversely to OBs. OCs co-cultured with CSC-spheres increase Ado production that is related to the OC resorption activity and contributes to T-cell suppression. Finally, we proved the efficacy of anti-CD73 agents in blocking NSCLC cell migration. Overall, we assessed the importance of adenosinergic signaling in the interaction between CSCs and OCs at the pre-metastatic niche, with therapeutic implications related to Ado production.
Collapse
|
19
|
Exogenous adenosine activates A2A adenosine receptor to inhibit RANKL-induced osteoclastogenesis via AP-1 pathway to facilitate bone repair. Mol Biol Rep 2021; 49:2003-2014. [PMID: 34846650 DOI: 10.1007/s11033-021-07017-1] [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: 09/07/2021] [Accepted: 11/24/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Adenosine is a purine nucleoside involved in regulating bone homeostasis through binding to A1, A2A, A2B, and A3 adenosine receptors (A1R, A2AR, A2BR, and A3R, respectively). However, the underlying mechanisms by which adenosine and receptor subtypes regulate osteoclast differentiation remain uncertain. This study aims to assess the role of exogenous adenosine and receptor subtypes in receptor activator of NF-κB ligand (RANKL)-induced osteoclast formation and explore the underlying molecular mechanisms. METHODS AND RESULTS The nanofibrous mats incorporated with adenosine exhibited robust ability to facilitate rat critical-size calvarial defect healing with decreased number of osteoclasts. Moreover, exogenous adenosine substantially enhanced the expression of A2AR and suppressed tartrate-resistant acid phosphatase-positive osteoclast formation and expression of osteoclast-related genes Ctsk, NFATc1, MMP9, and ACP5. This enhancement and suppression could be reversed by adding an A2AR antagonist, ZM241385, in RAW264.7 cells. Finally, RNA sequencing showed that the expression of Fos-related antigen 2 (Fra2) was distinctly downregulated through stimulation of adenosine in RAW264.7 cells treated with RANKL. This downregulation was reversed by ZM241385 according to real-time PCR, Western blot, and immunofluorescence analyses. CONCLUSIONS These findings demonstrated that exogenous adenosine binding to A2AR attenuated osteoclast differentiation via the inhibition of activating protein-1 (AP-1, including Fra2 subunit) pathway both in vitro and in vivo.
Collapse
|
20
|
Bone marrow CD73 + mesenchymal stem cells display increased stemness in vitro and promote fracture healing in vivo. Bone Rep 2021; 15:101133. [PMID: 34632004 PMCID: PMC8493579 DOI: 10.1016/j.bonr.2021.101133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/11/2021] [Accepted: 09/24/2021] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent and considered to be of great potential for regenerative medicine. We could show recently (Breitbach, Kimura et al. 2018) that a subpopulation of MSCs as well as sinusoidal endothelial cells (sECs) in the bone marrow (BM) of CD73-EGFP reporter mice could be labeled in vivo. We took advantage of this model to explore the plasticity and osteogenic potential of CD73-EGFP+ MSCs in vitro and their role in the regenerative response upon bone lesion in vivo. Herein we show that isolated CD73-EGFP+ MSCs displayed more pronounced stemness and stronger in vitro differentiation capacity into the osteogenic lineage compared to CD73-EGFP− MSCs. In a bone fracture model, endogenous BM-resident CD73-EGFP+ MSCs were found to migrate to the fracture site and differentiate into cartilage and bone cells. Our analysis also showed that CD73-EGFP+ sECs contributed to the neovascularization of the fracture site. In addition, grafting of CD73-EGFP+ MSCs into acute bone lesions revealed their capacity to differentiate into chondrocytes and osteocytes in vivo and their contribution to callus formation in the regeneration process of fracture healing. Thus, CD73+ MSCs display enhanced stemness and osteogenic differentiation potential in vitro and in vivo illustrating a prominent role of the CD73+ MSC subpopulation to promote fracture repair. CD73+ mesenchymal stem cells (MSCs) display pronounced proliferation potential. CD73+ MSCs show high in vitro osteogenic differentiation capacity. Endogenous bone marrow-resident CD73+ MSCs contribute to fracture healing. Grafted CD73+ MSCs promote bone formation upon fracture repair.
Collapse
|
21
|
Newman H, Shih YV, Varghese S. Resolution of inflammation in bone regeneration: From understandings to therapeutic applications. Biomaterials 2021; 277:121114. [PMID: 34488119 DOI: 10.1016/j.biomaterials.2021.121114] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/10/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022]
Abstract
Impaired bone healing occurs in 5-10% of cases following injury, leading to a significant economic and clinical impact. While an inflammatory response upon injury is necessary to facilitate healing, its resolution is critical for bone tissue repair as elevated acute or chronic inflammation is associated with impaired healing in patients and animal models. This process is governed by important crosstalk between immune cells through mediators that contribute to resolution of inflammation in the local healing environment. Approaches modulating the initial inflammatory phase followed by its resolution leads to a pro-regenerative environment for bone regeneration. In this review, we discuss the role of inflammation in bone repair, the negative impact of dysregulated inflammation on bone tissue regeneration, and how timely resolution of inflammation is necessary to achieve normal healing. We will discuss applications of biomaterials to treat large bone defects with a specific focus on resolution of inflammation to modulate the immune environment following bone injury, and their observed functional benefits. We conclude the review by discussing future strategies that could lead to the realization of anti-inflammatory therapeutics for bone tissue repair.
Collapse
Affiliation(s)
- Hunter Newman
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27710, USA
| | - Yuru Vernon Shih
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Shyni Varghese
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27710, USA; Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
22
|
Matthee C, Terre'Blanche G, Legoabe LJ, Janse van Rensburg HD. Exploration of chalcones and related heterocycle compounds as ligands of adenosine receptors: therapeutics development. Mol Divers 2021; 26:1779-1821. [PMID: 34176057 DOI: 10.1007/s11030-021-10257-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022]
Abstract
Adenosine receptors (ARs) are ubiquitously distributed throughout the mammalian body where they are involved in an extensive list of physiological and pathological processes that scientists have only begun to decipher. Resultantly, AR agonists and antagonists have been the focus of multiple drug design and development programmes within the past few decades. Considered to be a privileged scaffold in medicinal chemistry, the chalcone framework has attracted a substantial amount of interest in this regard. Due to the potential liabilities associated with its structure, however, it has become necessary to explore other potentially promising compounds, such as heterocycles, which have successfully been obtained from chalcone precursors in the past. This review aims to summarise the emerging therapeutic importance of adenosine receptors and their ligands, especially in the central nervous system (CNS), while highlighting chalcone and heterocyclic derivatives as promising AR ligand lead compounds.
Collapse
Affiliation(s)
- Chrisna Matthee
- Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa
| | - Gisella Terre'Blanche
- Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa.,Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa
| | - Lesetja J Legoabe
- Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa
| | - Helena D Janse van Rensburg
- Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa.
| |
Collapse
|
23
|
Verma NK, Kar AK, Singh A, Jagdale P, Satija NK, Ghosh D, Patnaik S. Control Release of Adenosine Potentiate Osteogenic Differentiation within a Bone Integrative EGCG- g-NOCC/Collagen Composite Scaffold toward Guided Bone Regeneration in a Critical-Sized Calvarial Defect. Biomacromolecules 2021; 22:3069-3083. [PMID: 34152738 DOI: 10.1021/acs.biomac.1c00513] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The regeneration of critical-sized bone defects with biomimetic scaffolds remains clinically challenging due to avascular necrosis, chronic inflammation, and altered osteogenic activity. Two confounding mechanisms, efficacy manipulation, and temporal regulation dictate the scaffold's bone regenerative ability. Equally critical is the priming of the mesenchymal stromal cells (MSCs) toward lineage-specific differentiation into bone-forming osteoblast, which particularly depends on varied mechanochemical and biological cues during bone tissue regeneration. This study sought to design and develop an optimized osteogenic scaffold, adenosine/epigallocatechin gallate-N,O-carboxymethyl chitosan/collagen type I (AD/EGCG-g-NOCC@clgn I), having osteoinductive components toward swift bone regeneration in a calvarial defect BALB/c mice model. The ex vivo findings distinctly establish the pro-osteogenic potential of adenosine and EGCG, stimulating MSCs toward osteoblast differentiation with significantly increased expression of alkaline phosphatase, calcium deposits, and enhanced osteocalcin expression. Moreover, the 3D matrix recapitulates extracellular matrix (ECM) properties, provides a favorable microenvironment, structural support against mechanical stress, and acts as a reservoir for sustained release of osteoinductive molecules for cell differentiation, proliferation, and migration during matrix osteointegration observed. Evidence from in vivo experiments, micro-CT analyses, histology, and histomorphometry signify accelerated osteogenesis both qualitatively and quantitatively: effectual bone union with enhanced bone formation and new ossified tissue in 4 mm sized defects. Our results suggest that the optimized scaffold serves as an adjuvant to guide bone tissue regeneration in critical-sized calvarial defects with promising therapeutic efficacy.
Collapse
Affiliation(s)
- Neeraj K Verma
- College of Dental Sciences, BBD University, Faizabad Road, Lucknow, Uttar Pradesh 226028, India
| | - Aditya K Kar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amrita Singh
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | | | - Neeraj K Satija
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debabrata Ghosh
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Satyakam Patnaik
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
24
|
Jauković A, Kukolj T, Trivanović D, Okić-Đorđević I, Obradović H, Miletić M, Petrović V, Mojsilović S, Bugarski D. Modulating stemness of mesenchymal stem cells from exfoliated deciduous and permanent teeth by IL-17 and bFGF. J Cell Physiol 2021; 236:7322-7341. [PMID: 33934350 DOI: 10.1002/jcp.30399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/12/2021] [Accepted: 04/05/2021] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) have been identified within dental pulp tissues of exfoliated deciduous (SHEDs) and permanent (DPSCs) teeth. Although differences in their proliferative and differentiation properties were revealed, variability in SHEDs and DPSCs responsiveness to growth factors and cytokines have not been studied before. Here, we investigated the influence of interleukin-17 (IL-17) and basic fibroblast growth factor (bFGF) on stemness features of SHEDs and DPSCs by analyzing their proliferation, clonogenicity, cell cycle progression, pluripotency markers expression and differentiation after 7-day treatment. Results indicated that IL-17 and bFGF differently affected SHEDs and DPSCs proliferation and clonogenicity, since bFGF increased proliferative and clonogenic potential of both cell types, while IL-17 similarly affected SHEDs, exerting no effects on adult counterparts DPSCs. In addition, both factors stimulated NANOG, OCT4, and SOX2 pluripotency markers expression in SHEDs and DPSCs showing diverse intracellular expression patterns dependent on MSCs type. As for the differentiation capacity, both factors displayed comparable effects on SHEDs and DPSCs, including stimulatory effect of IL-17 on early osteogenesis in contrast to the strong inhibitory effect showed for bFGF, while having no impact on SHEDs and DPSCs chondrogenesis. Moreover, bFGF combined with IL-17 reduced CD90 and stimulated CD73 expression on both types of MSCs, whereas each factor induced IL-6 expression indicating its' role in IL-17/bFGF-modulated properties of SHEDs and DPSCs. All these data demonstrated that dental pulp MSCs from primary and permanent teeth exert intrinsic features, providing novel evidence on how IL-17 and bFGF affect stem cell properties important for regeneration of dental pulp at different ages.
Collapse
Affiliation(s)
- Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Tamara Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Drenka Trivanović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia.,IZKF Research Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics, Würzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University Würzburg, Würzburg, Germany
| | - Ivana Okić-Đorđević
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Hristina Obradović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Maja Miletić
- Department of Pathophysiology, Faculty of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Vanja Petrović
- Department of Pediatric and Preventive Dentistry, Faculty of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Slavko Mojsilović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Diana Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
25
|
Zuccarini M, Giuliani P, Caciagli F, Ciccarelli R, Di Iorio P. In Search of a Role for Extracellular Purine Enzymes in Bone Function. Biomolecules 2021; 11:biom11050679. [PMID: 33946568 PMCID: PMC8147220 DOI: 10.3390/biom11050679] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Bone is one of the major tissues that undergoes continuous remodeling throughout life, thus ensuring both organic body growth during development and protection of internal organs as well as repair of trauma during adulthood. Many endogenous substances contribute to bone homeostasis, including purines. Their role has increasingly emerged in recent decades as compounds which, by interacting with specific receptors, can help determine adequate responses of bone cells to physiological or pathological stimuli. Equally, it is recognized that the activity of purines is closely dependent on their interconversion or metabolic degradation ensured by a series of enzymes present at extracellular level as predominantly bound to the cell membrane or, also, as soluble isoforms. While the effects of purines mediated by their receptor interactions have sufficiently, even though not entirely, been characterized in many tissues including bone, those promoted by the extracellular enzymes providing for purine metabolism have not been. In this review, we will try to circumstantiate the presence and the role of these enzymes in bone to define their close relationship with purine activities in maintaining bone homeostasis in normal or pathological conditions.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (P.G.); (P.D.I.)
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (P.G.); (P.D.I.)
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
| | - Francesco Caciagli
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (P.G.); (P.D.I.)
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
- StemTeCh Group, Via L. Polacchi, 66100 Chieti, Italy
- Correspondence:
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (P.G.); (P.D.I.)
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
| |
Collapse
|
26
|
Kar P, Millo T, Saha S, Mahtab S, Agarwal S, Goswami R. Osteogenic Mechanisms of Basal Ganglia Calcification and its ex vivo Model in the Hypoparathyroid Milieu. Endocrinology 2021; 162:6128830. [PMID: 33539507 DOI: 10.1210/endocr/bqab024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Indexed: 01/10/2023]
Abstract
CONTEXT Basal-ganglia calcification (BGC) is common (70%) in patients with chronic hypoparathyroidism. Interestingly, cortical gray matter is spared from calcification. The mechanism of BGC, role of hyperphosphatemia, and modulation of osteogenic molecules by parathyroid hormone (PTH) in its pathogenesis is not clear. OBJECTIVE We assessed the expression of a large repertoire of molecules with proosteogenic or antiosteogenic effects, including neuroprogenitor cells in caudate, dentate, and cortical gray matter from normal autopsy tissues. The effect of high phosphate and PTH was assessed in an ex vivo model of BGC using striatum tissue culture of the Sprague-Dawley rat. METHODS The messenger RNA and protein expression of 39 molecules involved in multiple osteogenic pathways were assessed in 25 autopsy tissues using reverse-transcriptase polymerase chain reaction, Western blot, and immunofluorescence. The striatal culture was maintained in a hypoparathyroid milieu for 24 days with and without (a) high phosphate (10-mm β-glycerophosphate) and (b) PTH(1-34) (50 ng/mL Dulbecco's modified Eagle's medium-F12 media) for their effect on striatal calcification and osteogenic molecules. RESULTS Procalcification molecules (osteonectin, β-catenin, klotho, FZD4, NT5E, LRP5, WNT3A, collagen-1α, and SOX2-positive neuroprogenitor stem cells) had significantly higher expression in the caudate than gray matter. Caudate nuclei also had higher expression of antiosteogenic molecules (osteopontin, carbonic anhydrase-II [CA-II], MGP, sclerostin, ISG15, ENPP1, and USP18). In an ex vivo model, striatum culture showed an increased propensity for calcified nodules with mineral deposition similar to that of bone tissue on Fourier-transformed infrared spectroscopy, alizarin, and von Kossa stain. Mineralization in striatal culture was enhanced by high phosphate and decreased by exogenous PTH through increased expression of CA-II. CONCLUSION This study provides a conceptual advance on the molecular mechanisms of BGC and the possibility of PTH therapy to prevent this complication in a hypoparathyroid milieu.
Collapse
Affiliation(s)
- Parmita Kar
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Tabin Millo
- Department of Forensic Medicine and Toxicology, New Delhi, Delhi, India
| | - Soma Saha
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Samrina Mahtab
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Shipra Agarwal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Ravinder Goswami
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, Delhi, India
| |
Collapse
|
27
|
Nakao Y, Fukuda T, Zhang Q, Sanui T, Shinjo T, Kou X, Chen C, Liu D, Watanabe Y, Hayashi C, Yamato H, Yotsumoto K, Tanaka U, Taketomi T, Uchiumi T, Le AD, Shi S, Nishimura F. Exosomes from TNF-α-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater 2021; 122:306-324. [PMID: 33359765 PMCID: PMC7897289 DOI: 10.1016/j.actbio.2020.12.046] [Citation(s) in RCA: 226] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/25/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cell (MSC)-derived exosome plays a central role in the cell-free therapeutics involving MSCs and the contents can be customized under disease-associated microenvironments. However, optimal MSC-preconditioning to enhance its therapeutic potential is largely unknown. Here, we show that preconditioning of gingival tissue-derived MSCs (GMSCs) with tumor necrosis factor-alpha (TNF-α) is ideal for the treatment of periodontitis. TNF-α stimulation not only increased the amount of exosome secreted from GMSCs, but also enhanced the exosomal expression of CD73, thereby inducing anti-inflammatory M2 macrophage polarization. The effect of GMSC-derived exosomes on inflammatory bone loss were examined by ligature-induced periodontitis model in mice. Local injection of GMSC-derived exosomes significantly reduced periodontal bone resorption and the number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts, and these effects were further enhanced by preconditioning of GMSCs with TNF-α. Thus, GMSC-derived exosomes also exhibited anti-osteoclastogenic activity. Receptor activator of NF-κB ligand (RANKL) expression was regulated by Wnt5a in periodontal ligament cells (PDLCs), and exosomal miR-1260b was found to target Wnt5a-mediated RANKL pathway and inhibit its osteoclastogenic activity. These results indicate that significant ability of the TNF-α-preconditioned GMSC-derived exosomes to regulate inflammation and osteoclastogenesis paves the way for establishment of a therapeutic approach for periodontitis.
Collapse
Affiliation(s)
- Yuki Nakao
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takao Fukuda
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan; Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, PA, USA
| | - Terukazu Sanui
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takanori Shinjo
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Xiaoxing Kou
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA; South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, China
| | - Chider Chen
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA; Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, PA, USA
| | - Dawei Liu
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA; Department of Orthodontics, Peking University School and Stomatology, Peking, China
| | - Yukari Watanabe
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Chikako Hayashi
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Hiroaki Yamato
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Karen Yotsumoto
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Urara Tanaka
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takaharu Taketomi
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Anh D Le
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, PA, USA
| | - Songtao Shi
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA; South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, China
| | - Fusanori Nishimura
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
28
|
Galgaro BC, Beckenkamp LR, van den M Nunnenkamp M, Korb VG, Naasani LIS, Roszek K, Wink MR. The adenosinergic pathway in mesenchymal stem cell fate and functions. Med Res Rev 2021; 41:2316-2349. [PMID: 33645857 DOI: 10.1002/med.21796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/02/2021] [Accepted: 02/17/2021] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) play an important role in tissue homeostasis and damage repair through their ability to differentiate into cells of different tissues, trophic support, and immunomodulation. These properties made them attractive for clinical applications in regenerative medicine, immune disorders, and cell transplantation. However, despite multiple preclinical and clinical studies demonstrating beneficial effects of MSCs, their native identity and mechanisms of action remain inconclusive. Since its discovery, the CD73/ecto-5'-nucleotidase is known as a classic marker for MSCs, but its role goes far beyond a phenotypic characterization antigen. CD73 contributes to adenosine production, therefore, is an essential component of purinergic signaling, a pathway composed of different nucleotides and nucleosides, which concentrations are finely regulated by the ectoenzymes and receptors. Thus, purinergic signaling controls pathophysiological functions such as proliferation, migration, cell fate, and immune responses. Despite the remarkable progress already achieved in considering adenosinergic pathway as a therapeutic target in different pathologies, its role is not fully explored in the context of the therapeutic functions of MSCs. Therefore, in this review, we provide an overview of the role of CD73 and adenosine-mediated signaling in the functions ascribed to MSCs, such as homing and proliferation, cell differentiation, and immunomodulation. Additionally, we will discuss the pathophysiological role of MSCs, via CD73 and adenosine, in different diseases, as well as in tumor development and progression. A better understanding of the adenosinergic pathway in the regulation of MSCs functions will help to provide improved therapeutic strategies applicable in regenerative medicine.
Collapse
Affiliation(s)
- Bruna C Galgaro
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Liziane R Beckenkamp
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Martha van den M Nunnenkamp
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Vitória G Korb
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Liliana I S Naasani
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Toruń, Poland
| | - Márcia R Wink
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
29
|
Extracellular purines and bone homeostasis. Biochem Pharmacol 2021; 187:114425. [PMID: 33482152 DOI: 10.1016/j.bcp.2021.114425] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
Maintenance of a healthy skeleton is highly dependent on an intricate regulation of bone metabolism, as changes in the balance between bone formation and bone resorption leads to bone loss, bone fragility and ultimately bone fractures. During the last three decades it has become increasingly evident that physiological release of purines in the extracellular space is imperative for bone homeostasis and is orchestrated via the network of purinoceptors. Adenosine derivatives are released locally in the skeleton either by the bone forming osteoblasts or the bone degrading osteoclasts actioned directly by processes like mechanical loading and indirectly by systemic hormones. Adenosine derivatives directly affect the bone cells by their action on the membranal receptors or have co-stimulatory actions with bone active hormones such as parathyroid hormone or the gut hormones. Any deviations leading to increased levels of extracellular adenosine derivatives in the bone tissue such as in pathologic situations, trigger complex pathways with opposing effects on tissue health as presented by studies involving a range of model organisms. Pathological conditions where skeletal purinergic signaling is affected are following tissue injury like microdamage and macroscopic fractures; and during inflammatory processes where nucleotides and nucleosides play an important part in the pathophysiological skeletal response. Moreover, adenosine derivatives also play an important role in the interaction between malignant cells and bone cells in several types of cancers involving the skeleton, such as but not limited to multiple myeloma and bone osteolysis. Much knowledge has been gained over the last decades. The net- resulting phenotype of adenosine derivatives in bone (including the ratio of ATP to Adenosine) is highly dependent on CD39 and CD73 enzymes together with the expression and activity of the specific receptors. Thus, each component is important in the physiological and pathophysiological processes in bone. Promising perspectives await in the future in treating skeletal disorders with medications targeting the individual components of the purinergic signaling pathway.
Collapse
|
30
|
Eisenstein A, Chitalia SV, Ravid K. Bone Marrow and Adipose Tissue Adenosine Receptors Effect on Osteogenesis and Adipogenesis. Int J Mol Sci 2020; 21:E7470. [PMID: 33050467 PMCID: PMC7589187 DOI: 10.3390/ijms21207470] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
Adenosine is an extracellular signaling molecule that is particularly relevant in times of cellular stress, inflammation and metabolic disturbances when the levels of the purine increase. Adenosine acts on two G-protein-coupled stimulatory and on two G-protein-coupled inhibitory receptors, which have varying expression profiles in different tissues and conditions, and have different affinities for the endogenous ligand. Studies point to significant roles of adenosine and its receptors in metabolic disease and bone health, implicating the receptors as potential therapeutic targets. This review will highlight our current understanding of the dichotomous effects of adenosine and its receptors on adipogenesis versus osteogenesis within the bone marrow to maintain bone health, as well as its relationship to obesity. Therapeutic implications will also be reviewed.
Collapse
Affiliation(s)
- Anna Eisenstein
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Shlok V. Chitalia
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA; (S.V.C.); (K.R.)
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA; (S.V.C.); (K.R.)
| |
Collapse
|
31
|
Allard B, Allard D, Buisseret L, Stagg J. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol 2020; 17:611-629. [PMID: 32514148 DOI: 10.1038/s41571-020-0382-2] [Citation(s) in RCA: 291] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2020] [Indexed: 12/14/2022]
Abstract
Cancer immunotherapy based on immune-checkpoint inhibition or adoptive cell therapy has revolutionized cancer care. Nevertheless, a large proportion of patients do not benefit from such treatments. Over the past decade, remarkable progress has been made in the development of 'next-generation' therapeutics in immuno-oncology, with inhibitors of extracellular adenosine (eADO) signalling constituting an expanding class of agents. Induced by tissue hypoxia, inflammation, tissue repair and specific oncogenic pathways, the adenosinergic axis is a broadly immunosuppressive pathway that regulates both innate and adaptive immune responses. Inhibition of eADO-generating enzymes and/or eADO receptors can promote antitumour immunity through multiple mechanisms, including enhancement of T cell and natural killer cell function, suppression of the pro-tumourigenic effects of myeloid cells and other immunoregulatory cells, and promotion of antigen presentation. With several clinical trials currently evaluating inhibitors of the eADO pathway in patients with cancer, we herein review the pathophysiological function of eADO with a focus on effects on antitumour immunity. We also discuss the treatment opportunities, potential limitations and biomarker-based strategies related to adenosine-targeted therapy in oncology.
Collapse
Affiliation(s)
- Bertrand Allard
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - David Allard
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Laurence Buisseret
- Department of Medical Oncology, Institut Jules Bordet, Brussels, Belgium
| | - John Stagg
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
32
|
Roccatello D, Fenoglio R, Sciascia S, Naretto C, Rossi D, Ferro M, Barreca A, Malavasi F, Baldovino S. CD38 and Anti-CD38 Monoclonal Antibodies in AL Amyloidosis: Targeting Plasma Cells and beyond. Int J Mol Sci 2020; 21:E4129. [PMID: 32531894 PMCID: PMC7312896 DOI: 10.3390/ijms21114129] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023] Open
Abstract
Immunoglobulin light chain amyloidosis (AL amyloidosis) is a rare systemic disease characterized by monoclonal light chains (LCs) depositing in tissue as insoluble fibrils, causing irreversible tissue damage. The mechanisms involved in aggregation and deposition of LCs are not fully understood, but CD138/38 plasma cells (PCs) are undoubtedly involved in monoclonal LC production.CD38 is a pleiotropic molecule detectable on the surface of PCs and maintained during the neoplastic transformation in multiple myeloma (MM). CD38 is expressed on T, B and NK cell populations as well, though at a lower cell surface density. CD38 is an ideal target in the management of PC dyscrasia, including AL amyloidosis, and indeed anti-CD38 monoclonal antibodies (MoAbs) have promising therapeutic potential. Anti-CD38 MoAbs act both as PC-depleting agents and as modulators of the balance of the immune cells. These aspects, together with their interaction with Fc receptors (FcRs) and neonatal FcRs, are specifically addressed in this paper. Moreover, the initiallyavailable experiences with the anti-CD38 MoAb DARA in AL amyloidosis are reviewed.
Collapse
Affiliation(s)
- Dario Roccatello
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy; (R.F.); (S.S.); (C.N.); (D.R.); (M.F.); (S.B.)
| | - Roberta Fenoglio
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy; (R.F.); (S.S.); (C.N.); (D.R.); (M.F.); (S.B.)
| | - Savino Sciascia
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy; (R.F.); (S.S.); (C.N.); (D.R.); (M.F.); (S.B.)
| | - Carla Naretto
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy; (R.F.); (S.S.); (C.N.); (D.R.); (M.F.); (S.B.)
| | - Daniela Rossi
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy; (R.F.); (S.S.); (C.N.); (D.R.); (M.F.); (S.B.)
| | - Michela Ferro
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy; (R.F.); (S.S.); (C.N.); (D.R.); (M.F.); (S.B.)
| | - Antonella Barreca
- Pathology Division, Department of Oncology, University of Turin, 10154 Turin, Italy;
| | - Fabio Malavasi
- Department of Medical Science, University of Turin, and Fondazione Ricerca Molinette, 10154 Turin, Italy;
| | - Simone Baldovino
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy; (R.F.); (S.S.); (C.N.); (D.R.); (M.F.); (S.B.)
| |
Collapse
|
33
|
Chandrasekaran B, Samarneh S, Jaber AMY, Kassab G, Agrawal N. Therapeutic Potentials of A2B Adenosine Receptor Ligands: Current Status and Perspectives. Curr Pharm Des 2020; 25:2741-2771. [PMID: 31333084 DOI: 10.2174/1381612825666190717105834] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Adenosine receptors (ARs) are classified as A1, A2A, A2B, and A3 subtypes belong to the superfamily of G-protein coupled receptors (GPCRs). More than 40% of modern medicines act through either activation or inhibition of signaling processes associated with GPCRs. In particular, A2B AR signaling pathways are implicated in asthma, inflammation, cancer, ischemic hyperfusion, diabetes mellitus, cardiovascular diseases, gastrointestinal disorders, and kidney disease. METHODS This article reviews different disease segments wherein A2B AR is implicated and discusses the potential role of subtype-selective A2B AR ligands in the management of such diseases or disorders. All the relevant publications on this topic are reviewed and presented scientifically. RESULTS This review provides an up-to-date highlight of the recent advances in the development of novel and selective A2B AR ligands and their therapeutic role in treating various disease conditions. A special focus has been given to the therapeutic potentials of selective A2B AR ligands in the management of airway inflammatory conditions and cancer. CONCLUSIONS This systematic review demonstrates the current status and perspectives of A2B AR ligands as therapeutically useful agents that would assist medicinal chemists and pharmacologists in discovering novel and subtype-selective A2B AR ligands as potential drug candidates.
Collapse
Affiliation(s)
- Balakumar Chandrasekaran
- Faculty of Pharmacy, Philadelphia University-Jordan, P. O. Box: 1, Philadelphia University-19392, Amman, Jordan
| | - Sara Samarneh
- Faculty of Pharmacy, Philadelphia University-Jordan, P. O. Box: 1, Philadelphia University-19392, Amman, Jordan
| | - Abdul Muttaleb Yousef Jaber
- Faculty of Pharmacy, Philadelphia University-Jordan, P. O. Box: 1, Philadelphia University-19392, Amman, Jordan
| | - Ghadir Kassab
- Faculty of Pharmacy, Philadelphia University-Jordan, P. O. Box: 1, Philadelphia University-19392, Amman, Jordan
| | - Nikhil Agrawal
- College of Health Sciences, University of KwaZulu-Natal, P. O. Box: 4000, Westville, Durban, South Africa
| |
Collapse
|
34
|
Wang Y, Copeland J, Shin M, Chang Y, Venton BJ. CD73 or CD39 Deletion Reveals Different Mechanisms of Formation for Spontaneous and Mechanically Stimulated Adenosine and Sex Specific Compensations in ATP Degradation. ACS Chem Neurosci 2020; 11:919-928. [PMID: 32083837 DOI: 10.1021/acschemneuro.9b00620] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adenosine is important for local neuromodulation, and rapid adenosine signaling can occur spontaneously or after mechanical stimulation, but little is known about how adenosine is formed in the extracellular space for those stimulations. Here, we studied mechanically stimulated and spontaneous adenosine to determine if rapid adenosine is formed by extracellular breakdown of adenosine triphosphate (ATP) using mice globally deficient in extracellular breakdown enzymes, either CD39 (nucleoside triphosphate diphosphohydrolase 1, NTPDase1) or CD73 (ecto-5'-nucleotidase). CD39 knockout (KO) mice have a lower frequency of spontaneous adenosine events than wild-type (WT, C57BL/6). Surprisingly, CD73KO mice demonstrate sex differences in spontaneous adenosine; males maintain similar event frequencies as WT, but females have significantly fewer events and lower concentrations. Examining the mRNA expression of other enzymes that metabolize ATP revealed tissue nonspecific alkaline phosphatase (TNAP) was upregulated in male CD73KO mice, but not secreted prostatic acid phosphatase (PAP) or transmembrane PAP. Thus, TNAP upregulation compensates for CD73 loss in males but not in females. These sex differences highlight that spontaneous adenosine is formed by metabolism of extracellular ATP by many enzymes. For mechanically stimulated adenosine, CD39KO or CD73KO did not change stimulation frequency, concentration, or t1/2. Thus, the mechanism of formation for mechanically stimulated adenosine is likely direct release of adenosine, different than spontaneous adenosine. Understanding these different mechanisms of rapid adenosine formation will help to develop pharmacological treatments that differentially target modes of rapid adenosine signaling, and all treatments should be studied in both sexes, given possible differences in extracellular ATP degradation.
Collapse
Affiliation(s)
- Ying Wang
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Jeffrey Copeland
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Biology, Eastern Mennonite University, Harrisonburg, Virginia 22802, United States
| | - Mimi Shin
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Yuanyu Chang
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - B. Jill Venton
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
35
|
Corciulo C, Cronstein BN. Signaling of the Purinergic System in the Joint. Front Pharmacol 2020; 10:1591. [PMID: 32038258 PMCID: PMC6993121 DOI: 10.3389/fphar.2019.01591] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
The joint is a complex anatomical structure consisting of different tissues, each with a particular feature, playing together to give mobility and stability at the body. All the joints have a similar composition including cartilage for reducing the friction of the movement and protecting the underlying bone, a synovial membrane that produces synovial fluid to lubricate the joint, ligaments to limit joint movement, and tendons for the interaction with muscles. Direct or indirect damage of one or more of the tissues forming the joint is the foundation of different pathological conditions. Many molecular mechanisms are involved in maintaining the joint homeostasis as well as in triggering disease development. The molecular pathway activated by the purinergic system is one of them.The purinergic signaling defines a group of receptors and intermembrane channels activated by adenosine, adenosine diphosphate, adenosine 5’-triphosphate, uridine triphosphate, and uridine diphosphate. It has been largely described as a modulator of many physiological and pathological conditions including rheumatic diseases. Here we will give an overview of the purinergic system in the joint describing its expression and function in the synovium, cartilage, ligament, tendon, and bone with a therapeutic perspective.
Collapse
Affiliation(s)
- Carmen Corciulo
- Division of Translational Medicine, Department of Medicine, NYU School of Medicine, New York, NY, United States.,Krefting Research Centre-Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Bruce N Cronstein
- Division of Translational Medicine, Department of Medicine, NYU School of Medicine, New York, NY, United States.,Division of Rheumatology, Department of Medicine, NYU School of Medicine, New York, NY, United States
| |
Collapse
|
36
|
Pinto-Cardoso R, Pereira-Costa F, Pedro Faria J, Bandarrinha P, Bessa-Andrês C, Correia-de-Sá P, Bernardo Noronha-Matos J. Adenosinergic signalling in chondrogenesis and cartilage homeostasis: Friend or foe? Biochem Pharmacol 2019; 174:113784. [PMID: 31884043 DOI: 10.1016/j.bcp.2019.113784] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Chondrocytes and their mesenchymal cell progenitors secrete a variety of bioactive molecules, including adenine nucleotides and nucleosides, but these molecules are not usually highlighted in review papers about the secretome of these cells. Ageing and inflammatory insults compromise chondrocytes ability to keep ATP/adenosine synthesis, release and turnover. Cartilage homeostasis depends on extracellular adenosine levels, which acting via four P1 purinoceptor subtypes modulates the release of pro-inflammatory mediators, including NO, PGE2 and several cytokines. Native articular cartilage is challenged by synovial fluid flow during normal joint motion transiently increasing ATP release and adenosine formation in the joint microenvironment. Excessive joint motion and shockwave trauma are deleterious to cartilage homeostasis due to HIF-1α overexpression, resulting in disproportionate ecto-5'-nucleotidase/CD73 production, adenosine accumulation and superfluous A2B receptors activation. Scarcity of data however exists on the putative interplay between coexistent high affinity (A2A and A3) and low affinity (A2B) adenosine receptors activation affecting stem cells fate towards preferential chondrogenic or osteogenic lineages in the human cartilage. Hints gathered in this commentary result mainly from studies using human immortalized cell lines and animal (e.g. rodent, equine, bovine) tissue samples. The available data point towards adenosine A2A and A3 receptors having cartilage protective roles, while excessive adenosine accumulation may be detrimental via low affinity A2B receptors activation, with little reference to the putative role of the adenosine forming enzyme ecto-5'-nucleotidase/CD73. Thus, emphasizing the multiple pathways responsible for controlling adenosine signalling in cartilage will certainly impact on the search for novel therapeutic targets for highly disabling articular disorders.
Collapse
Affiliation(s)
- Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - Flávio Pereira-Costa
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - João Pedro Faria
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - Patrícia Bandarrinha
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal.
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Portugal.
| |
Collapse
|
37
|
Shih YRV, Liu M, Kwon SK, Iida M, Gong Y, Sangaj N, Varghese S. Dysregulation of ectonucleotidase-mediated extracellular adenosine during postmenopausal bone loss. SCIENCE ADVANCES 2019; 5:eaax1387. [PMID: 31457100 PMCID: PMC6703860 DOI: 10.1126/sciadv.aax1387] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/12/2019] [Indexed: 05/25/2023]
Abstract
Adenosine and its receptors play a key role in bone homeostasis and regeneration. Extracellular adenosine is generated from CD39 and CD73 activity in the cell membrane, through conversion of adenosine triphosphate to adenosine monophosphate (AMP) and AMP to adenosine, respectively. Despite the relevance of CD39/CD73 to bone health, the roles of these enzymes in bona fide skeletal disorders remain unknown. We demonstrate that CD39/CD73 expression and extracellular adenosine levels in the bone marrow are substantially decreased in animals with osteoporotic bone loss. Knockdown of estrogen receptors ESR1 and ESR2 in primary osteoprogenitors and osteoclasts undergoing differentiation showed decreased coexpression of membrane-bound CD39 and CD73 and lower extracellular adenosine. Targeting the adenosine A2B receptor using an agonist attenuated bone loss in ovariectomized mice. Together, these findings suggest a pathological association of purine metabolism with estrogen deficiency and highlight the potential of A2B receptor as a target to treat osteoporosis.
Collapse
Affiliation(s)
- Yu-Ru V. Shih
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Mengqian Liu
- Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Seong Keun Kwon
- Department of Otorhinolaryngology–Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | | | - Ya Gong
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nivedita Sangaj
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
- Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
38
|
Giuliani N, Accardi F, Marchica V, Dalla Palma B, Storti P, Toscani D, Vicario E, Malavasi F. Novel targets for the treatment of relapsing multiple myeloma. Expert Rev Hematol 2019; 12:481-496. [PMID: 31125526 DOI: 10.1080/17474086.2019.1624158] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Introduction: Multiple myeloma (MM) is characterized by the high tendency to relapse and develop drug resistance. Areas covered: This review focused on the main novel targets identified to design drugs for the treatment of relapsing MM patients. CD38 and SLAMF7 are the main surface molecules leading to the development of monoclonal antibodies (mAbs) recently approved for the treatment of relapsing MM patients. B cell maturation antigen (BCMA) is a suitable target for antibody-drug conjugates, bispecific T cell engager mAbs and Chimeric Antigen Receptor (CAR)-T cells. Moreover, the programmed cell death protein 1 (PD)-1/PD-Ligand (PD-L1) expression profile by MM cells and their microenvironment and the use of immune checkpoints inhibitors in MM patients are reported. Finally, the role of histone deacetylase (HDAC), B cell lymphoma (BCL)-2 family proteins and the nuclear transport protein exportin 1 (XPO1) as novel targets are also underlined. The clinical results of the new inhibitors in relapsing MM patients are discussed. Expert opinion: CD38, SLAMF7, and BCMA are the main targets for different immunotherapeutic approaches. Selective inhibitors of HDAC6, BCL-2, and XPO1 are new promising compounds under clinical investigation in relapsing MM patients.
Collapse
Affiliation(s)
- Nicola Giuliani
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Fabrizio Accardi
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Valentina Marchica
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | | | - Paola Storti
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Denise Toscani
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Emanuela Vicario
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Fabio Malavasi
- b Department of Medical Science , University of Turin , Turin , Italy
| |
Collapse
|
39
|
Avdeeva E, Shults E, Rybalova T, Reshetov Y, Porokhova E, Sukhodolo I, Litvinova L, Shupletsova V, Khaziakhmatova O, Khlusov I, Guryev A, Belousov M. Chelidonic Acid and Its Derivatives from Saussurea Controversa: Isolation, Structural Elucidation and Influence on the Osteogenic Differentiation of Multipotent Mesenchymal Stromal Cells In Vitro. Biomolecules 2019; 9:biom9050189. [PMID: 31100934 PMCID: PMC6572306 DOI: 10.3390/biom9050189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/02/2019] [Accepted: 05/13/2019] [Indexed: 01/18/2023] Open
Abstract
4-oxo-4H-pyran-2.6-dicarboxylic acid (chelidonic acid, ChA) in the native state and in the complex with calcium [Ca(ChA)(H2O)3], named saucalchelin (CaChA), was isolated from the extract of Saussurea controversa leaves for the first time for the Asteraceae family. The structure of ChA was determined by NMR, MS and confirmed by X-ray analysis of its monomethyl ester, and CaChA was described by IR, ICP-MS, CHN analysis. The yield of ChA and CaChA was 45 mg/g and 70 mg/g of extract, respectively. The osteogenic activity of ChA, n-monobutyl ester of chelidonic acid, and CaChA has been studied in vitro in a 21-day culture of human adipose-derived multipotent mesenchymal stromal cells (hAMMSCs) in a standard nutrient medium without osteogenic supplements. CaChA significantly stimulated the growth of cell mass and differentiation of hAMMSCs into osteoblasts with subsequent mineralization of the culture and it may be a promising substance for accelerating bone tissue regeneration and engineering.
Collapse
Affiliation(s)
- Elena Avdeeva
- Department of Pharmaceutical Analysis, Siberian State Medical University, Tomsk 634050, Russia.
| | - Elvira Shults
- Laboratory of Medicinal Chemistry, Novosibirsk Institute of Organic Chemistry, Siberian Branch, Novosibirsk 630090, Russia.
- Novosibirsk State University, 2 Pirogova St., Novosibirsk 630090, Russia.
| | - Tatyana Rybalova
- Novosibirsk State University, 2 Pirogova St., Novosibirsk 630090, Russia.
- Center of Spectral Investigations, Novosibirsk Institute of Organic Chemistry, Siberian Branch, Novosibirsk 630090, Russia.
| | - Yaroslav Reshetov
- Department of Pharmaceutical Analysis, Siberian State Medical University, Tomsk 634050, Russia.
| | - Ekaterina Porokhova
- Department of Morphology and General Pathology, Siberian State Medical University, Tomsk 634050, Russia.
| | - Irina Sukhodolo
- Department of Morphology and General Pathology, Siberian State Medical University, Tomsk 634050, Russia.
| | - Larisa Litvinova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia.
| | - Valeria Shupletsova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia.
| | - Olga Khaziakhmatova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia.
| | - Igor Khlusov
- Department of Morphology and General Pathology, Siberian State Medical University, Tomsk 634050, Russia.
- Research School of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia.
| | - Artem Guryev
- Department of Pharmaceutical Analysis, Siberian State Medical University, Tomsk 634050, Russia.
| | - Mikhail Belousov
- Department of Pharmaceutical Analysis, Siberian State Medical University, Tomsk 634050, Russia.
- Research School of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia.
| |
Collapse
|
40
|
Noda S, Kawashima N, Yamamoto M, Hashimoto K, Nara K, Sekiya I, Okiji T. Effect of cell culture density on dental pulp-derived mesenchymal stem cells with reference to osteogenic differentiation. Sci Rep 2019; 9:5430. [PMID: 30931957 PMCID: PMC6443725 DOI: 10.1038/s41598-019-41741-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 03/11/2019] [Indexed: 01/09/2023] Open
Abstract
Dental pulp stem cells (DPSCs) are a good source for tissue regeneration, however, the number of DPSCs in the pulp tissue is limited. Cell propagation is essential for tissue engineering using DPSCs and the cell culture conditions may affect the properties of DPSCs. The purpose of this study was to analyze the effect of cell culture condition, especially dense culture condition, on the property and differentiation pathway of DPSCs. We cultured DPSCs under sparse (sDPSCs; 5 × 103 cells/cm2) or dense (dDPSCs; 1 × 105 cells/cm2) conditions for 4 days and compared their properties. The populations of CD73+ and CD105+ cells were significantly decreased in dDPSCs. Both groups showed multi-differentiation potential, but mineralized nodule formation was enhanced in dDPSCs. The phosphorylation of focal adhesion kinase (FAK) and phosphoinositide 3-kinase (PI3K) proteins was promoted in dDPSCs, and alkaline phosphatase (ALP) mRNA expression in dDPSCs was abolished in the presence of pan-PI3K and FAK inhibitors. dDPSCs implanted into mouse bone cavities induced more mineralized tissue formation than sDPSCs and control. These findings indicate that dense culture conditions modified the properties of DPSCs and gave rise to osteogenic-lineage commitment via integrin signaling and suggest that dense culture conditions favor the propagation of DPSCs to be used for mineralized tissue regeneration.
Collapse
Affiliation(s)
- Sonoko Noda
- Department of Pulp Biology and Endodontics, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Nobuyuki Kawashima
- Department of Pulp Biology and Endodontics, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan.
| | - Mioko Yamamoto
- Department of Pulp Biology and Endodontics, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Kentaro Hashimoto
- Department of Pulp Biology and Endodontics, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Keisuke Nara
- Department of Pulp Biology and Endodontics, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Takashi Okiji
- Department of Pulp Biology and Endodontics, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| |
Collapse
|
41
|
Ottensmeyer PF, Witzler M, Schulze M, Tobiasch E. Small Molecules Enhance Scaffold-Based Bone Grafts via Purinergic Receptor Signaling in Stem Cells. Int J Mol Sci 2018; 19:E3601. [PMID: 30441872 PMCID: PMC6274752 DOI: 10.3390/ijms19113601] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022] Open
Abstract
The need for bone grafts is high, due to age-related diseases, such as tumor resections, but also accidents, risky sports, and military conflicts. The gold standard for bone grafting is the use of autografts from the iliac crest, but the limited amount of accessible material demands new sources of bone replacement. The use of mesenchymal stem cells or their descendant cells, namely osteoblast, the bone-building cells and endothelial cells for angiogenesis, combined with artificial scaffolds, is a new approach. Mesenchymal stem cells (MSCs) can be obtained from the patient themselves, or from donors, as they barely cause an immune response in the recipient. However, MSCs never fully differentiate in vitro which might lead to unwanted effects in vivo. Interestingly, purinergic receptors can positively influence the differentiation of both osteoblasts and endothelial cells, using specific artificial ligands. An overview is given on purinergic receptor signaling in the most-needed cell types involved in bone metabolism-namely osteoblasts, osteoclasts, and endothelial cells. Furthermore, different types of scaffolds and their production methods will be elucidated. Finally, recent patents on scaffold materials, as wells as purinergic receptor-influencing molecules which might impact bone grafting, are discussed.
Collapse
Affiliation(s)
- Patrick Frank Ottensmeyer
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Markus Witzler
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| |
Collapse
|
42
|
Wang Y, Kim J, Chan A, Whyne C, Nam D. A two phase regulation of bone regeneration: IL-17F mediates osteoblastogenesis via C/EBP-β in vitro. Bone 2018; 116:47-57. [PMID: 30010083 DOI: 10.1016/j.bone.2018.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/28/2018] [Accepted: 07/10/2018] [Indexed: 01/15/2023]
Abstract
T lymphocytes and pro-inflammatory cytokines, specifically interleukin-17F (IL-17F) have been identified as important regulators in bone regeneration during fracture repair. To better understand the molecular mechanisms of IL-17F-mediated osteoblastogenesis, a mouse pre-osteoblast cell line (MC3T3-E1) was utilized to characterize the intracellular signal transduction of IL-17F. Comparisons to the established canonical Wnt signaling pathway were made using Wnt3a ligand. Our results demonstrated greater bone marker gene expression in IL-17F-treated cells, compared to cells treated with Wnt3a. Western blot analysis confirmed degradation of β-catenin and up-regulation of two key proteins in osteoblast differentiation, Runx2 and C/EBP-β, in response to IL-17F treatment. RNA silencing of IL-17F receptors, IL-17Ra and IL-17Rc via siRNA transfection resulted in decreased expression of Act2, Runx2, and C/EBP-β, demonstrating the direct ligand-receptor interaction between IL-17F and IL-17Ra/c as an activator of osteoblastogenesis. Our findings suggest that IL-17F promotes osteoblast differentiation independent of the canonical Wnt pathway and β-catenin signaling, presenting new insights on modulating the adaptive immune response in the inflammatory phase, temporally distinct from the reparative and remodeling phases of fracture healing.
Collapse
Affiliation(s)
- Yufa Wang
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Jieun Kim
- MD/PhD Program, University of Toronto, Toronto, ON, Canada
| | - Andrea Chan
- Division of Orthopaedic Surgery, University of Toronto, Toronto, ON, Canada
| | - Cari Whyne
- Sunnybrook Research Institute, Toronto, ON, Canada; Division of Orthopaedic Surgery, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada
| | - Diane Nam
- Sunnybrook Research Institute, Toronto, ON, Canada; Division of Orthopaedic Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW To summarize and discuss recent progress and novel signaling mechanisms relevant to bone marrow adipocyte formation and its physiological/pathophysiological implications for bone remodeling. RECENT FINDINGS Skeletal remodeling is a coordinated process entailing removal of old bone and formation of new bone. Several bone loss disorders such as osteoporosis are commonly associated with increased bone marrow adipose tissue. Experimental and clinical evidence supports that a reduction in osteoblastogenesis from mesenchymal stem cells at the expense of adipogenesis, as well as the deleterious effects of adipocyte-derived signaling, contributes to the etiology of osteoporosis as well as bone loss associated with aging, diabetes mellitus, post-menopause, and chronic drug therapy. However, this view is challenged by findings indicating that, in some contexts, bone marrow adipose tissue may have a beneficial impact on skeletal health. Further research is needed to better define the role of marrow adipocytes in bone physiology/pathophysiology and to determine the therapeutic potential of manipulating mesenchymal stem cell differentiation.
Collapse
Affiliation(s)
- Shanmugam Muruganandan
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA
| | - Christopher J Sinal
- Department of Pharmacology, Dalhousie University, 5850 College Street, Box 15000, Halifax, Nova Scotia, B3H4R2, Canada.
| |
Collapse
|
44
|
Fas-L promotes the stem cell potency of adipose-derived mesenchymal cells. Cell Death Dis 2018; 9:695. [PMID: 29891848 PMCID: PMC5995957 DOI: 10.1038/s41419-018-0702-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/26/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
Abstract
Fas-L is a TNF family member known to trigger cell death. It has recently become evident that Fas-L can transduce also non-apoptotic signals. Mesenchymal stem cells (MSCs) are multipotent cells that are derived from various adult tissues. Although MSCs from different tissues display common properties they also display tissue-specific characteristics. Previous works have demonstrated massive apoptosis following Fas-L treatment of bone marrow-derived MSCs both in vitro and following their administration in vivo. We therefore set to examine Fas-L-induced responses in adipose-derived stem cells (ASCs). Human ASCs were isolated from lipoaspirates and their reactivity to Fas-L treatment was examined. ASCs responded to Fas-L by simultaneous apoptosis and proliferation, which yielded a net doubling of cell quantities and a phenotypic shift, including reduced expression of CD105 and increased expression of CD73, in association with increased bone differentiation potential. Treatment of freshly isolated ASCs led to an increase in large colony forming unit fibroblasts, likely produced by early stem cell progenitor cells. Fas-L-induced apoptosis and proliferation signaling were found to be independent as caspase inhibition attenuated Fas-L-induced apoptosis without impacting proliferation, whereas inhibition of PI3K and MEK, but not of JNK, attenuated Fas-L-dependent proliferation, but not apoptosis. Thus, Fas-L signaling in ASCs leads to their expansion and phenotypic shift toward a more potent stem cell state. We speculate that these reactions ensure the survival of ASC progenitor cells encountering Fas-L-enriched environments during tissue damage and inflammation and may also enhance ASC survival following their administration in vivo.
Collapse
|
45
|
Tangjit N, Dechkunakorn S, Anuwongnukroh N, Khaneungthong A, Sritanaudomchai H. Optimal Xeno-free Culture Condition for Clinical Grade Stem Cells from Human Exfoliated Deciduous Teeth. Int J Stem Cells 2018; 11:96-104. [PMID: 29482310 PMCID: PMC5984063 DOI: 10.15283/ijsc17049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 11/14/2017] [Accepted: 01/25/2018] [Indexed: 01/06/2023] Open
Abstract
Background and Objectives Stem cells from human exfoliated deciduous teeth (SHED) are a promising clinical resource for various tissue defects, including lumbar spondylosis, neural compression, and cleft palate. Use of media containing animal-derived serum carries potential risk of infectious diseases and unwanted immunogenicity. To increase the potential utility of SHED for clinical application, SHED was adapted to xeno-free conditions. Methods Define xeno-free culture media were compared with the conventional serum containing media in the culture of SHED. Cultured SHED in different media were further characterized through proliferative capacities, cellular phenotype, and differentiation potential. Results Selected xeno-free media were capable of supporting the growth of SHED. MSCGM-CD Bulletkit medium greatly increased the number and proliferate capacity of colony-forming unit-fibroblast than SHED cultured in other media. In addition, the characteristic surface markers expression and multipotent differentiation potential of SHED in the MSCGM-CD Bulletkit medium were comparable to those observed with serum-containing medium. Conclusions The xeno-free medium described herein has the potential to be further used for the safe expansion and to determine efficient way to produce clinical grade dental stem cells for therapeutic applications.
Collapse
Affiliation(s)
- Nathaphon Tangjit
- Department of Orthodontics, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Surachai Dechkunakorn
- Department of Orthodontics, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Niwat Anuwongnukroh
- Department of Orthodontics, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Anuson Khaneungthong
- Department of Orthodontics, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
46
|
Corciulo C, Irrera N, Cronstein BN. Adenosine Receptors Regulate Bone Remodeling and Cartilage Physiology. THE ADENOSINE RECEPTORS 2018:515-527. [DOI: 10.1007/978-3-319-90808-3_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
47
|
Feig JL, Mediero A, Corciulo C, Liu H, Zhang J, Perez-Aso M, Picard L, Wilder T, Cronstein B. The antiviral drug tenofovir, an inhibitor of Pannexin-1-mediated ATP release, prevents liver and skin fibrosis by downregulating adenosine levels in the liver and skin. PLoS One 2017; 12:e0188135. [PMID: 29145453 PMCID: PMC5690602 DOI: 10.1371/journal.pone.0188135] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022] Open
Abstract
Background Fibrosing diseases are a leading cause of morbidity and mortality worldwide and, therefore, there is a need for safe and effective antifibrotic therapies. Adenosine, generated extracellularly by the dephosphorylation of adenine nucleotides, ligates specific receptors which play a critical role in development of hepatic and dermal fibrosis. Results of recent clinical trials indicate that tenofovir, a widely used antiviral agent, reverses hepatic fibrosis/cirrhosis in patients with chronic hepatitis B infection. Belonging to the class of acyclic nucleoside phosphonates, tenofovir is an analogue of AMP. We tested the hypothesis that tenofovir has direct antifibrotic effects in vivo by interfering with adenosine pathways of fibrosis using two distinct models of adenosine and A2AR-mediated fibrosis. Methods Thioacetamide (100mg/kg IP)-treated mice were treated with vehicle, or tenofovir (75mg/kg, SubQ) (n = 5–10). Bleomycin (0.25U, SubQ)-treated mice were treated with vehicle or tenofovir (75mg/kg, IP) (n = 5–10). Adenosine levels were determined by HPLC, and ATP release was quantitated as luciferase-dependent bioluminescence. Skin breaking strength was analysed and H&E and picrosirus red-stained slides were imaged. Pannexin-1expression was knocked down following retroviral-mediated expression of of Pannexin-1-specific or scrambled siRNA. Results Treatment of mice with tenofovir diminished adenosine release from the skin of bleomycin-treated mice and the liver of thioacetamide-treated mice, models of diffuse skin fibrosis and hepatic cirrhosis, respectively. More importantly, tenofovir treatment diminished skin and liver fibrosis in these models. Tenofovir diminished extracellular adenosine concentrations by inhibiting, in a dose-dependent fashion, cellular ATP release but not in cells lacking Pannexin-1. Conclusions These studies suggest that tenofovir, a widely used antiviral agent, could be useful in the treatment of fibrosing diseases.
Collapse
Affiliation(s)
- Jessica L. Feig
- Division of Translational Medicine, Department of Medicine, NYU-Langone Medical Center, New York, New York, United States of America
| | - Aranzazu Mediero
- Division of Translational Medicine, Department of Medicine, NYU-Langone Medical Center, New York, New York, United States of America
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Carmen Corciulo
- Division of Translational Medicine, Department of Medicine, NYU-Langone Medical Center, New York, New York, United States of America
| | - Hailing Liu
- Division of Translational Medicine, Department of Medicine, NYU-Langone Medical Center, New York, New York, United States of America
| | - Jin Zhang
- Division of Translational Medicine, Department of Medicine, NYU-Langone Medical Center, New York, New York, United States of America
- Department of Immunology and Rheumatology, LiHuili Hospital, Medical School of Ningbo University, Ningbo, China
| | - Miguel Perez-Aso
- Division of Translational Medicine, Department of Medicine, NYU-Langone Medical Center, New York, New York, United States of America
| | - Laura Picard
- Division of Translational Medicine, Department of Medicine, NYU-Langone Medical Center, New York, New York, United States of America
| | - Tuere Wilder
- Division of Translational Medicine, Department of Medicine, NYU-Langone Medical Center, New York, New York, United States of America
| | - Bruce Cronstein
- Division of Translational Medicine, Department of Medicine, NYU-Langone Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
48
|
Sobreiro-Almeida R, Tamaño-Machiavello MN, Carvalho EO, Cordón L, Doria S, Senent L, Correia DM, Ribeiro C, Lanceros-Méndez S, Sabater I Serra R, Gomez Ribelles JL, Sempere A. Human Mesenchymal Stem Cells Growth and Osteogenic Differentiation on Piezoelectric Poly(vinylidene fluoride) Microsphere Substrates. Int J Mol Sci 2017; 18:ijms18112391. [PMID: 29137121 PMCID: PMC5713360 DOI: 10.3390/ijms18112391] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/22/2022] Open
Abstract
The aim of this work was to determine the influence of the biomaterial environment on human mesenchymal stem cell (hMSC) fate when cultured in supports with varying topography. Poly(vinylidene fluoride) (PVDF) culture supports were prepared with structures ranging between 2D and 3D, based on PVDF films on which PVDF microspheres were deposited with varying surface density. Maintenance of multipotentiality when cultured in expansion medium was studied by flow cytometry monitoring the expression of characteristic hMSCs markers, and revealed that cells were losing their characteristic surface markers on these supports. Cell morphology was assessed by scanning electron microscopy (SEM). Alkaline phosphatase activity was also assessed after seven days of culture on expansion medium. On the other hand, osteoblastic differentiation was monitored while culturing in osteogenic medium after cells reached confluence. Osteocalcin immunocytochemistry and alizarin red assays were performed. We show that flow cytometry is a suitable technique for the study of the differentiation of hMSC seeded onto biomaterials, giving a quantitative reliable analysis of hMSC-associated markers. We also show that electrosprayed piezoelectric poly(vinylidene fluoride) is a suitable support for tissue engineering purposes, as hMSCs can proliferate, be viable and undergo osteogenic differentiation when chemically stimulated.
Collapse
Affiliation(s)
- R Sobreiro-Almeida
- Centro/Departamento de Física, Universidade do Minho, 4710-057 Braga, Portugal.
| | - M N Tamaño-Machiavello
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain.
| | - E O Carvalho
- Centro/Departamento de Física, Universidade do Minho, 4710-057 Braga, Portugal.
| | - L Cordón
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain.
| | - S Doria
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain.
| | - L Senent
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain.
- Hematology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain.
| | - D M Correia
- Centro/Departamento de Física, Universidade do Minho, 4710-057 Braga, Portugal.
- Centro/Departamento de Química, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| | - C Ribeiro
- Centro/Departamento de Física, Universidade do Minho, 4710-057 Braga, Portugal.
- CEB-Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| | - S Lanceros-Méndez
- BCMaterials, Parque Científico y Tecnológico de Bizkaia, 48160 Derio, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
| | - R Sabater I Serra
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain.
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 46022 Valencia, Spain.
| | - J L Gomez Ribelles
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain.
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 46022 Valencia, Spain.
| | - A Sempere
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain.
- Hematology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain.
| |
Collapse
|
49
|
Bradaschia-Correa V, Josephson AM, Egol AJ, Mizrahi MM, Leclerc K, Huo J, Cronstein BN, Leucht P. Ecto-5'-nucleotidase (CD73) regulates bone formation and remodeling during intramembranous bone repair in aging mice. Tissue Cell 2017; 49:545-551. [PMID: 28720305 PMCID: PMC5656528 DOI: 10.1016/j.tice.2017.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 07/03/2017] [Accepted: 07/03/2017] [Indexed: 11/19/2022]
Abstract
Ecto-5'-nucleotidase (CD73) generates adenosine, an osteoblast activator and key regulator of skeletal growth. It is unknown, however, if CD73 regulates osteogenic differentiation during fracture healing in adulthood, and in particular how CD73 activity regulates intramembranous bone repair in the elderly. Monocortical tibial defects were created in 46-52-week-old wild type (WT) and CD73 knock-out mice (CD73-/-) mice. Injury repair was analyzed at post-operative days 5, 7, 14 and 21 by micro-computed tomography (micro-CT), histomorphometry, proliferating cell nuclear antigen (PCNA) immunostaining, alkaline phosphatase (ALP) and tartrate-resistant acid phosphatase (TRAP) histochemistry. Middle-aged CD73 knock-out mice exhibited delayed bone regeneration and significantly reduced bone matrix deposition detected by histomorphometry and micro-CT. Cell proliferation, ALP activity and osteoclast number were reduced in the CD73-/- mice, suggesting a combined defect in bone formation and resorption due the absence of CD73 activity in this model of intramembranous bone repair. Results from this study demonstrate that osteoblast activation through CD73 activity is essential during bone repair in aging mice, and it may present a drugable target for future biomimetic therapeutic approaches that aim at enhancing bone formation in the elderly patients.
Collapse
Affiliation(s)
- Vivian Bradaschia-Correa
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Anne M Josephson
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Alexander J Egol
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Matthew M Mizrahi
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Kevin Leclerc
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Jason Huo
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Bruce N Cronstein
- Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Philipp Leucht
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA; Department of Cell Biology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
50
|
Hau KL, Ranzoni AM, Vlahova F, Hawkins K, De Coppi P, David AL, Guillot PV. TGFβ-induced osteogenic potential of human amniotic fluid stem cells via CD73-generated adenosine production. Sci Rep 2017; 7:6601. [PMID: 28747757 PMCID: PMC5529586 DOI: 10.1038/s41598-017-06780-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 06/16/2017] [Indexed: 12/22/2022] Open
Abstract
The human amniotic fluid stem cell (hAFSC) population consists of two morphologically distinct subtypes, spindle-shaped and round-shaped cells (SS-hAFSCs and RS-hAFSCs). Whilst SS-hAFSCs are routinely expanded in mesenchymal-type (MT) conditions, we previously showed that they acquire broader differentiation potential when cultured under embryonic-type (ET) conditions. However, the effects of culture conditions on RS-hAFSCs have not been determined. Here, we show that culturing RS-hAFSCs under ET conditions confers faster proliferation and enhances the efficiency of osteogenic differentiation of the cells. We show that this occurs via TGFβ-induced activation of CD73 and the associated increase in the generation of extracellular adenosine. Our data demonstrate that culture conditions are decisive for the expansion of hAFSCs and that TGFβ present in ET conditions causes the phenotype of RS-hAFSCs to revert to an earlier state of stemness. Cultivating RS-hAFSCs in ET conditions with TGFβ may therefore increase their therapeutic potential for clinical applications.
Collapse
Affiliation(s)
- Kwan-Leong Hau
- National Heart & Lung Institute, Hammersmith Campus, Du Cane Road, Imperial College London, London, W12 0NN, UK.,Institute for Women's Health, Research Department of Maternal and Fetal Medicine, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Anna Maria Ranzoni
- Institute for Women's Health, Research Department of Maternal and Fetal Medicine, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Filipa Vlahova
- Institute for Women's Health, Research Department of Maternal and Fetal Medicine, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Kate Hawkins
- Institute for Women's Health, Research Department of Maternal and Fetal Medicine, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Paolo De Coppi
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Anna L David
- Institute for Women's Health, Research Department of Maternal and Fetal Medicine, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Pascale V Guillot
- Institute for Women's Health, Research Department of Maternal and Fetal Medicine, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|