1
|
Reyes Fernandez PC, Wright CS, Farach-Carson MC, Thompson WR. Examining Mechanisms for Voltage-Sensitive Calcium Channel-Mediated Secretion Events in Bone Cells. Calcif Tissue Int 2023; 113:126-142. [PMID: 37261463 PMCID: PMC11008533 DOI: 10.1007/s00223-023-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
In addition to their well-described functions in cell excitability, voltage-sensitive calcium channels (VSCCs) serve a critical role in calcium (Ca2+)-mediated secretion of pleiotropic paracrine and endocrine factors, including those produced in bone. Influx of Ca2+ through VSCCs activates intracellular signaling pathways to modulate a variety of cellular processes that include cell proliferation, differentiation, and bone adaptation in response to mechanical stimuli. Less well understood is the role of VSCCs in the control of bone and calcium homeostasis mediated through secreted factors. In this review, we discuss the various functions of VSCCs in skeletal cells as regulators of Ca2+ dynamics and detail how these channels might control the release of bioactive factors from bone cells. Because VSCCs are druggable, a better understanding of the multiple functions of these channels in the skeleton offers the opportunity for developing new therapies to enhance and maintain bone and to improve systemic health.
Collapse
Affiliation(s)
- Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX, 77005, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
2
|
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
|
3
|
Fu Y, Li C, Wang Q, Gao R, Cai X, Wang S, Zhang Y. The protective effect of collagen peptides from bigeye tuna (Thunnus obesus) skin and bone to attenuate UVB-induced photoaging via MAPK and TGF-β signaling pathways. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
4
|
Wright CS, Robling AG, Farach-Carson MC, Thompson WR. Skeletal Functions of Voltage Sensitive Calcium Channels. Curr Osteoporos Rep 2021; 19:206-221. [PMID: 33721180 PMCID: PMC8216424 DOI: 10.1007/s11914-020-00647-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 12/15/2022]
Abstract
Voltage-sensitive calcium channels (VSCCs) are ubiquitous multimeric protein complexes that are necessary for the regulation of numerous physiological processes. VSCCs regulate calcium influx and various intracellular processes including muscle contraction, neurotransmission, hormone secretion, and gene transcription, with function specificity defined by the channel's subunits and tissue location. The functions of VSCCs in bone are often overlooked since bone is not considered an electrically excitable tissue. However, skeletal homeostasis and adaptation relies heavily on VSCCs. Inhibition or deletion of VSCCs decreases osteogenesis, impairs skeletal structure, and impedes anabolic responses to mechanical loading. RECENT FINDINGS: While the functions of VSCCs in osteoclasts are less clear, VSCCs have distinct but complementary functions in osteoblasts and osteocytes. PURPOSE OF REVIEW: This review details the structure, function, and nomenclature of VSCCs, followed by a comprehensive description of the known functions of VSCCs in bone cells and their regulation of bone development, bone formation, and mechanotransduction.
Collapse
Affiliation(s)
- Christian S Wright
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Alexander G Robling
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy & Cell Biology, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic & Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, 77054, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy & Cell Biology, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
5
|
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: 3.7] [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
|
6
|
Zhou Y, Arredondo HM, Wang N. P2Y Receptors in Bone - Anabolic, Catabolic, or Both? Front Endocrinol (Lausanne) 2021; 12:818499. [PMID: 35069456 PMCID: PMC8777008 DOI: 10.3389/fendo.2021.818499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/17/2021] [Indexed: 11/13/2022] Open
Abstract
P2Y receptors, including eight subtypes, are G protein-coupled receptors that can be activated by extracellular nucleotides. Nearly all P2Y receptors are expressed in bone cells, suggesting their involvements in bone physiology and pathology. However, their exact roles in bone homeostasis are not entirely clear. Therefore, this mini review summarizes new research developments regarding individual P2Y receptors and their roles in bone biology, particularly detailing those which execute both anabolic and catabolic functions. This dual function has highlighted the conundrum of pharmacologically targeting these P2Y receptors in bone-wasting diseases. Further research in finding more precise targeting strategy, such as promoting anabolic effects via combining with physical exercise, should be prioritized.
Collapse
|
7
|
Hioki T, Tokuda H, Nakashima D, Fujita K, Kawabata T, Sakai G, Kim W, Tachi J, Tanabe K, Matsushima-Nishiwaki R, Otsuka T, Iida H, Kozawa O. HSP90 inhibitors strengthen extracellular ATP-stimulated synthesis of interleukin-6 in osteoblasts: Amplification of p38 MAP kinase. Cell Biochem Funct 2020; 39:88-97. [PMID: 32567086 DOI: 10.1002/cbf.3566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/03/2020] [Accepted: 05/17/2020] [Indexed: 12/22/2022]
Abstract
Heat shock protein 90 (HSP90) is expressed ubiquitously in a variety of cell types including osteoblasts. HSP90 acts as a key driver of proteostasis under pathophysiological conditions. Here, we investigated the involvement of HSP90 in extracellular ATP-stimulated interleukin (IL)-6 synthesis and HSP90 downstream signalling in osteoblast-like MC3T3-E1 cells. In osteoblasts, extracellular ATP stimulates the synthesis of IL-6, a bone-remodelling agent. Geldanamycin, 17-allylamino-17-demethoxy-geldanamycin (17-AAG) and onalespib, three different HSP90 inhibitors, amplified the ATP-stimulated IL-6 release. Geldanamycin increased IL-6 mRNA expression elicited by ATP. ATP enhanced the triiodothyronine-induced osteocalcin release, but HSP90 inhibitors suppressed the release. Extracellular ATP induced the phosphorylation of p44/p42 mitogen-activated protein kinase (MAPK), p38 MAPK, c-Jun N-terminal kinase (JNK), p70 S6 kinase, Akt, and myosin phosphatase-targeting subunit (MYPT), a Rho-kinase substrate. SB203580, an inhibitor of p38 MAPK, suppressed ATP-stimulated IL-6 release. Inhibitors of MEK1/2 (PD98059), JNK (SP600125), upstream kinase of p70 S6 kinase (rapamycin) and Akt (deguelin), all increased IL-6 release. Y27632, a Rho-kinase inhibitor, failed to affect the IL-6 release stimulated by ATP. Geldanamycin and 17-AAG both amplified ATP-induced p38 MAPK phosphorylation, although geldanamycin inhibited the phosphorylation of Akt induced by ATP. In addition, SB203580 significantly reduced the amplification by geldanamycin of the IL-6 release. Taken together, our results strongly suggest that HSP90 inhibitors up-regulate extracellular ATP-stimulated IL-6 synthesis via amplification of p38 MAPK activation in osteoblasts. SIGNIFICANCE OF THE STUDY: Heat shock protein 90 (HSP90) acts as a key driver of proteostasis under pathophysiological conditions in a variety of cell types. We have previously shown that HSP90 is expressed at high levels in osteoblast-like MC3T3-E1 cells, even in their quiescent state, consistent with HSP90 performing an important physiological function in osteoblasts. In the present study, we investigated whether HSP90 is implicated in extracellular ATP-induced interleukin (IL)-6 synthesis in osteoblast-like MC3T3-E1 cells. Our results strongly suggest that HSP90 inhibitors up-regulate extracellular ATP-stimulated IL-6 synthesis via amplification of p38 mitogen-activated protein kinase activation in osteoblasts.
Collapse
Affiliation(s)
- Tomoyuki Hioki
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Dermatology, Kizawa Memorial Hospital, Minokamo, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Clinical Laboratory/Biobank of Medical Genome Centre, National Centre for Geriatrics and Gerontology, Obu, Japan
| | - Daiki Nakashima
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Anaesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kazuhiko Fujita
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tetsu Kawabata
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Go Sakai
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Woo Kim
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Anaesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Junko Tachi
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Anaesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kumiko Tanabe
- Department of Anaesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | | | - Takanobu Otsuka
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroki Iida
- Department of Anaesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
8
|
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: 2.5] [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
|
9
|
Hao Q, Liu Z, Lu L, Zhang L, Zuo L. Both JNK1 and JNK2 Are Indispensable for Sensitized Extracellular Matrix Mineralization in IKKβ-Deficient Osteoblasts. Front Endocrinol (Lausanne) 2020; 11:13. [PMID: 32117051 PMCID: PMC7028708 DOI: 10.3389/fendo.2020.00013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/08/2020] [Indexed: 01/05/2023] Open
Abstract
Extracellular matrix mineralization is critical for osteogenesis, and its dysregulation could result in osteoporosis and vascular calcification. IKK/NF-κB activation inhibits differentiation of osteoblasts, and reduces extracellular matrix mineralization, however the underlying mechanisms are poorly understood. In this study, we used CRISPR/Cas9 system to permanently inactivate IKKβ in preosteoblast cells and confirmed that such cells displayed dramatic increase in extracellular matrix mineralization associated with JNK phosphorylation. Such observation was also found in our study using IKKβ-deficient primary murine osteoblasts. Interestingly, we found that in Ikbkb-/-Mapk8-/- or Ikbkb-/-Mapk9-/- double knockout cells, the enhanced mineralization caused by IKKβ deficiency was completely abolished, and deletion of either Mapk8 or Mapk9 was sufficient to dampen c-Jun phosphorylation. In further experiments, we discovered that absence of JNK1 or JNK2 on IKKβ-deficient background resulted in highly conserved transcriptomic alteration in response to osteogenic induction. Therefore, identification of the indispensable roles of JNK1 and JNK2 in activating c-Jun and promoting osteoblast differentiation on IKKβ-deficient background provided novel insights into restoring homeostasis in extracellular matrix mineralization.
Collapse
Affiliation(s)
- Qianyun Hao
- Department of Nephrology, Peking University People's Hospital, Beijing, China
| | - Zhuangzhuang Liu
- Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Liaoxun Lu
- Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Lichen Zhang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Lichen Zhang
| | - Li Zuo
- Department of Nephrology, Peking University People's Hospital, Beijing, China
- *Correspondence: Li Zuo
| |
Collapse
|
10
|
Du D, Zhou Z, Zhu L, Hu X, Lu J, Shi C, Chen F, Chen A. TNF-α suppresses osteogenic differentiation of MSCs by accelerating P2Y 2 receptor in estrogen-deficiency induced osteoporosis. Bone 2018; 117:161-170. [PMID: 30236554 DOI: 10.1016/j.bone.2018.09.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/13/2018] [Accepted: 09/16/2018] [Indexed: 12/12/2022]
Abstract
Tumor Necrosis Factor-α (TNF-α)-inhibited osteogenic differentiation of mesenchymal stem cells (MSCs) contributes to impaired bone formation, which plays a central role in the pathogenesis of postmenopausal osteoporosis. However, the exact mechanisms of TNF-α-inhibited osteoblast differentiation have not been fully elucidated. Multiple P2 purinoceptor subtypes are expressed in several species of osteoblasts and are confirmed to regulate bone metabolism. The purpose of this study is to investigate whether P2 purinoceptors are involved in TNF-α-inhibited osteoblast differentiation. This study shows TNF-α increased P2Y2 receptor expression in the differentiation of MSCs into osteoblasts in a noticeable manner. Overexpressing or silencing of the P2Y2 receptor either impaired or promoted osteogenic differentiation of MSCs significantly. Silencing of the P2Y2 receptor attenuated the inhibitory effects of TNF-α on osteoblastic differentiation of MSCs. In addition, silencing of the P2Y2 receptor evidently alleviated TNF-α-inhibited MSC proliferation. P2Y2 receptor expression was mechanistically upregulated by TNF-α mainly through extracellular regulated protein kinase (ERK) and c-Jun N-terminal kinase (JNK) signaling pathways. Overall, our results revealed a novel function of the P2Y2 receptor and suggested suppressing the P2Y2 receptor may be an effective strategy to promote bone formation in estrogen deficiency-induced osteoporosis.
Collapse
Affiliation(s)
- Di Du
- Department of Orthopedics and Trauma Surgery, Changzheng Hospital, the Second Military Medical University, Shanghai, China
| | - Zhibin Zhou
- Department of Orthopedics and Trauma Surgery, Changzheng Hospital, the Second Military Medical University, Shanghai, China
| | - Lei Zhu
- Department of Orthopedics and Trauma Surgery, Changzheng Hospital, the Second Military Medical University, Shanghai, China
| | - Xianteng Hu
- Department of Orthopedics and Trauma Surgery, Changzheng Hospital, the Second Military Medical University, Shanghai, China
| | - Jiajia Lu
- Department of Orthopedics and Trauma Surgery, Changzheng Hospital, the Second Military Medical University, Shanghai, China
| | - Changgui Shi
- Department of Spine Surgery, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Fangjing Chen
- Department of Orthopedics, General Hospital of Jinan Military Command, Jinan 250031, Shandong, China.
| | - Aimin Chen
- Department of Orthopedics and Trauma Surgery, Changzheng Hospital, the Second Military Medical University, Shanghai, China.
| |
Collapse
|
11
|
Roszek K, Wujak M. How to influence the mesenchymal stem cells fate? Emerging role of ectoenzymes metabolizing nucleotides. J Cell Physiol 2018; 234:320-334. [PMID: 30078187 DOI: 10.1002/jcp.26904] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 06/13/2018] [Indexed: 12/11/2022]
Abstract
Extracellular purines, principally adenosine triphosphate and adenosine, are among the oldest evolutionary and widespread chemical messengers. The integrative view of purinergic signaling as a multistage coordinated cascade involves the participation of nucleotides/nucleosides, their receptors, enzymes metabolizing extracellular nucleosides and nucleotides as well as several membrane transporters taking part in the release and/or uptake of these molecules. In view of the emerging data, it is evident and widely accepted that an extensive network of diverse enzymatic activities exists in the extracellular space. The enzymes regulate the availability of nucleotide and adenosine receptor agonists, and consequently, the course of signaling events. The current data indicate that mesenchymal stem cells (MSCs) and cells induced to differentiate exhibit different sensitivity to purinergic ligands as well as a distinct activity and expression profiles of ectonucleotidases than mature cells. In the proposed review, we postulate for a critical role of these enzymatic players which, by orchestrating a fine-tune regulation of nucleotides concentrations, are integrally involved in modulation and diversification of purinergic signals. This specific hallmark of the MSC purinome should be linked with cell-specific biological potential and capacity for tissue regeneration. We anticipate this publication to be a starting point for scientific discussion and novel approach to the in vitro and in vivo regulation of the MSC properties.
Collapse
Affiliation(s)
- Katarzyna Roszek
- Biochemistry Department, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Magdalena Wujak
- Biochemistry Department, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
12
|
Laiuppa JA, Santillán GE. Involvement of GSK3/β-catenin in the action of extracellular ATP on differentiation of primary cultures from rat calvaria into osteoblasts. J Cell Biochem 2018; 119:8378-8388. [PMID: 29932242 DOI: 10.1002/jcb.27037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/05/2018] [Indexed: 11/08/2022]
Abstract
Modulation of purinergic receptors play an important role in the regulation of osteoblasts differentiation and bone formation. In this study, we investigated the involvement of the GSK3/βcatenin signaling in the action of ATPγ-S on osteogenic differentiation of primary cell cultures from rat calvaria. Our results indicate that the cell treatment with 10 or 100 µM ATPγ-S for 96 h increase the cytoplasmic levels of β-catenin and its translocation to nucleus respect to control. A similar effect was observed after cell treatment with the GSK3 inhibitor LiCl (10 mM). Cell treatments with 4-10 mM LiCl significantly stimulated ALP activity respect to control at 4 and 7 days, suggesting that inhibition of GSK-3 mediates osteoblastic differentiation of rat calvarial cells. Effects comparison between ATP and LiCl shown that ALP activity was significantly increased by 10 µM ATPγ-S and decreased by 10 mM LiCl at 10 day of treatment, respect to control, suggesting that the effect of ATPγ-S was less potent but more persistent than of LiCl in stimulating this osteogenic marker in calvarial cells. Cell culture mineralization was significantly increased by treatment with 10 µM ATPγ-S and decreased by 10 mM LiCl, respect to control. In together, these results suggest that GSK3 inhibition is involved in ATPγ-S action on rat calvarial cell differentiation into osteoblasts at early steadies. In addition such inhibition by LiCl appear promote osteoblasts differentiation at beginning but has a deleterious effect on its function at later steadies as the extracellular matrix mineralization.
Collapse
Affiliation(s)
- Juan A Laiuppa
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, INBIOSUR-CONICET, Bahía Blanca, Argentina
| | - Graciela E Santillán
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, INBIOSUR-CONICET, Bahía Blanca, Argentina
| |
Collapse
|
13
|
Xu Y, Shu B, Tian Y, Chelly M, Morandi MM, Barton S, Shang X, Dong Y. Notch activation promotes osteoblast mineralization by inhibition of apoptosis. J Cell Physiol 2018; 233:6921-6928. [PMID: 29693255 DOI: 10.1002/jcp.26592] [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] [Received: 12/03/2017] [Accepted: 03/09/2018] [Indexed: 12/18/2022]
Abstract
Notch activator Jagged1 (JAG1) plays a critical role in the regulation of osteoblast differentiation and bone metabolism. In this study, JAG1-induced osteoblast proliferation, differentiation, and mineralization has been analyzed in primary osteoblasts for up to 7 days. Alkaline phosphatase and Alizarin red staining showed an enhanced osteoblast maturation and mineralization in JAG1 treated cells, as well as higher mRNA levels of late osteoblast differentiation markers. In contrast, Notch inhibitor DAPT and deletion of Runx2 totally blocked JAG1 effects on osteoblast mineralization. Flow cytometry data further showed a significantly higher cell proliferation in early stages of culture at day 3, and lower levels of osteoblast apoptosis in late stages of culture at day 7. More importantly, activation of anti-apoptotic factor BCL-2 was enhanced, while pro-apoptotic factor Caspase3 was reduced in JAG1 treated osteoblasts. Therefore, we conclude that cell mineralization is enhanced via anti-apoptotic actions of Notch signaling within the osteoblast cells.
Collapse
Affiliation(s)
- Ying Xu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Bing Shu
- Spine Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ye Tian
- Department of Orthopaedic Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Marjorie Chelly
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Massimo M Morandi
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Shane Barton
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Xifu Shang
- Department of Orthopaedic Surgery, Anhui Provincial Hospital, Hefei, Anhui, China
| | - Yufeng Dong
- Spine Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| |
Collapse
|
14
|
Orriss IR, Guneri D, Hajjawi MOR, Shaw K, Patel JJ, Arnett TR. Activation of the P2Y 2 receptor regulates bone cell function by enhancing ATP release. J Endocrinol 2017; 233:341-356. [PMID: 28420708 DOI: 10.1530/joe-17-0042] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 04/18/2017] [Indexed: 02/02/2023]
Abstract
Bone cells constitutively release ATP into the extracellular environment where it acts locally via P2 receptors to regulate bone cell function. Whilst P2Y2 receptor stimulation regulates bone mineralisation, the functional effects of this receptor in osteoclasts remain unknown. This investigation used the P2Y2 receptor knockout (P2Y2R-/- ) mouse model to investigate the role of this receptor in bone. MicroCT analysis of P2Y2R-/- mice demonstrated age-related increases in trabecular bone volume (≤48%), number (≤30%) and thickness (≤17%). In vitro P2Y2R-/- osteoblasts displayed a 3-fold increase in bone formation and alkaline phosphatase activity, whilst P2Y2R-/- osteoclasts exhibited a 65% reduction in resorptive activity. Serum cross-linked C-telopeptide levels (CTX, resorption marker) were also decreased (≤35%). The resorption defect in P2Y2R-/- osteoclasts was rescued by the addition of exogenous ATP, suggesting that an ATP deficit could be a key factor in the reduced function of these cells. In agreement, we found that basal ATP release was reduced up to 53% in P2Y2R-/- osteoclasts. The P2Y2 receptor agonists, UTP and 2-thioUTP, increased osteoclast activity and ATP release in wild-type but not in P2Y2R-/- cells. This indicates that the P2Y2 receptor may regulate osteoclast function indirectly by promoting ATP release. UTP and 2-thioUTP also stimulate ATP release from osteoblasts suggesting that the P2Y2 receptor exerts a similar function in these cells. Taken together, our findings are consistent with the notion that the primary action of P2Y2 receptor signalling in bone is to regulate extracellular ATP levels.
Collapse
Affiliation(s)
- Isabel R Orriss
- Department of Comparative Biomedical SciencesRoyal Veterinary College, London, UK
| | - Dilek Guneri
- Department of Comparative Biomedical SciencesRoyal Veterinary College, London, UK
| | - Mark O R Hajjawi
- Department of Cell & Developmental BiologyUniversity College London, London, UK
| | - Kristy Shaw
- Department of Comparative Biomedical SciencesRoyal Veterinary College, London, UK
| | - Jessal J Patel
- Department of Comparative Biomedical SciencesRoyal Veterinary College, London, UK
| | - Timothy R Arnett
- Department of Cell & Developmental BiologyUniversity College London, London, UK
| |
Collapse
|
15
|
Anguita E, Villalobo A. Src-family tyrosine kinases and the Ca 2+ signal. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:915-932. [PMID: 27818271 DOI: 10.1016/j.bbamcr.2016.10.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/25/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023]
Abstract
In this review, we shall describe the rich crosstalk between non-receptor Src-family kinases (SFKs) and the Ca2+ transient generated in activated cells by a variety of extracellular and intracellular stimuli, resulting in diverse signaling events. The exchange of information between SFKs and Ca2+ is reciprocal, as it flows in both directions. These kinases are main actors in pathways leading to the generation of the Ca2+ signal, and reciprocally, the Ca2+ signal modulates SFKs activity and functions. We will cover how SFKs participate in the generation of the cytosolic Ca2+ rise upon activation of a series of receptors and the mechanism of clearance of this Ca2+ signal. The role of SFKs modulating Ca2+-translocating channels participating in these events will be amply discussed. Finally, the role of the Ca2+ sensor protein calmodulin on the activity of c-Src, and potentially on other SFKs, will be outlined as well. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Estefanía Anguita
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
16
|
Gofman L, Fernandes NC, Potula R. Relative Role of Akt, ERK and CREB in Alcohol-Induced Microglia P2X4R Receptor Expression. Alcohol Alcohol 2016; 51:647-654. [PMID: 26946194 PMCID: PMC5091293 DOI: 10.1093/alcalc/agw009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 02/05/2016] [Accepted: 02/09/2016] [Indexed: 12/12/2022] Open
Abstract
AIMS Previously we have demonstrated altered microglia P2X4R expression in response to alcohol and pharmacological blockade with a selective P2X4R antagonist can reverse the action, suggesting that P2X4R play a role in mediating alcohol-induced effects on microglia. In the present study, we investigated the underlying signaling mediators, which may play a role in modulating P2X4R expression in microglia cells in response to alcohol. METHODS Embryonic stem cell-derived microglia (ESdM) cells were used to investigate the potential mechanisms involved in the regulation of P2X4R in response to alcohol. Selective P2X4R antagonist and kinase inhibitors were used to further corroborate the signal transduction pathway through which alcohol modulates P2X4R expression in microglia. RESULTS Alcohol (100 mM) suppressed phosphorylated AKT and ERK cascades in native ESdM cells. This alcohol-induced suppression was confirmed to be P2X4R-dependent through the use of a selective P2X4R antagonist and knockdown of P2XR4 by siRNA. Alcohol increased transcriptional activity of CREB. P2X4R antagonist blocked alcohol-induced effects on CREB, suggesting a P2X4R-mediated effect. CONCLUSION These findings provide important clues to the underlying mechanism of purinoceptors in alcohol-induced microglia immune suppression.
Collapse
Affiliation(s)
- Larisa Gofman
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Nicole C Fernandes
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Raghava Potula
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
17
|
Cutarelli A, Marini M, Tancredi V, D'Arcangelo G, Murdocca M, Frank C, Tarantino U. Adenosine Triphosphate stimulates differentiation and mineralization in human osteoblast-like Saos-2 cells. Dev Growth Differ 2016; 58:400-8. [PMID: 27189526 DOI: 10.1111/dgd.12288] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 03/16/2016] [Accepted: 03/22/2016] [Indexed: 12/25/2022]
Abstract
In the last years adenosine triphosphate (ATP) and subsequent purinergic system activation through P2 receptors were investigated highlighting their pivotal role in bone tissue biology. In osteoblasts ATP can regulate several activities like cell proliferation, cell death, cell differentiation and matrix mineralization. Since controversial results exist, in this study we analyzed the ATP effects on differentiation and mineralization in human osteoblast-like Saos-2 cells. We showed for the first time the altered functional activity of ATP receptors. Despite that, we found that ATP can reduce cell proliferation and stimulate osteogenic differentiation mainly in the early stages of in vitro maturation as evidenced by the enhanced expression of alkaline phosphatase (ALP), Runt-related transcription factor 2 (Runx2) and Osteocalcin (OC) genes and by the increased ALP activity. Moreover, we found that ATP can affect mineralization in a biphasic manner, at low concentrations ATP always increases mineral deposition while at high concentrations it always reduces mineral deposition. In conclusion, we show the osteogenic effect of ATP on both early and late stage activities like differentiation and mineralization, for the first time in human osteoblastic cells.
Collapse
Affiliation(s)
- Alessandro Cutarelli
- Department of Orthopaedics and Traumatology, University Hospital Foundation, Policlinico Tor Vergata, Viale Oxford 81, Rome, 00133, Italy.,National Centre for Rare Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy
| | - Mario Marini
- Department of Medicine of Systems, University of Rome Tor Vergata, Via Montpellier 1, Italy
| | - Virginia Tancredi
- Department of Medicine of Systems, University of Rome Tor Vergata, Via Montpellier 1, Italy
| | - Giovanna D'Arcangelo
- Department of Medicine of Systems, University of Rome Tor Vergata, Via Montpellier 1, Italy
| | - Michela Murdocca
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, Rome, 00133, Italy
| | - Claudio Frank
- National Centre for Rare Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy
| | - Umberto Tarantino
- Department of Orthopaedics and Traumatology, University Hospital Foundation, Policlinico Tor Vergata, Viale Oxford 81, Rome, 00133, Italy
| |
Collapse
|
18
|
Laiuppa JA, Santillán GE. Effect of Combined Action of Extracellular ATP and Elevated Calcium on Osteogenic Differentiation of Primary Cultures From Rat Calvaria. J Cell Biochem 2016; 117:2658-68. [PMID: 27038365 DOI: 10.1002/jcb.25565] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/31/2016] [Indexed: 11/06/2022]
Abstract
The in vitro osteogenic differentiation has been intensively studied. However, it is not yet clear precisely how osteogenesis can be optimized. Changes in extracellular Ca(2+) concentration ([Ca(2+) ]e ), as well as modulation of purinergic receptors play an important role in the regulation of osteoblasts differentiation and bone formation. In this study, we investigated the effects of a combined treatment of ATPγ-S and high [Ca(2+) ]e (5.35 mM) on osteogenic differentiation and function of primary cell cultures from rat calvaria. Our results indicate that ATPγ-S stimulates cell transition from the G0 to S phase of cell cycle, involving the PI3K signaling pathway. Treatment with 10 or 100 µM ATPγ-S and [Ca(2+) ]e (ATP-[Ca(2+) ]e ) for 48 h increases cell number significantly above the control. ATPγ-S treatment in osteogenic medium containing [Ca(2+) ]e stimulates the gene expression of BMP-4, BMP-5, and OPN at 16, 48, and 72 h, respectively, above control. In same conditions, treatment for 6 days with 10 µM UTP or 100 µM UDP significantly increased the ALP activity respect to control. Cells grown in osteogenic medium showed a statistically significant increase in calcium deposits at 15 and 18 days, for 10 µM ATPγ-S treatment, and at 18 and 22 days, for [Ca(2+) ]e treatment, respect to control but ATP-[Ca(2+) ]e treatment shown a significant greater mineralization at 15 days respect to ATPγ-S, and at 18 days respect to both agonists. In conclusion, we demonstrated that an osteogenic medium containing 10 µM ATPγ-S and 5.35 mM [Ca(2+) ]e enhance osteogenesis and mineralization by rat primary calvarial cells cultures. J. Cell. Biochem. 117: 2658-2668, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Juan A Laiuppa
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, CONICET, San Juan 670, (B8000ICN) Bahía Blanca, Argentina
| | - Graciela E Santillán
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, CONICET, San Juan 670, (B8000ICN) Bahía Blanca, Argentina.
| |
Collapse
|
19
|
Noronha-Matos JB, Correia-de-Sá P. Mesenchymal Stem Cells Ageing: Targeting the "Purinome" to Promote Osteogenic Differentiation and Bone Repair. J Cell Physiol 2016; 231:1852-61. [PMID: 26754327 DOI: 10.1002/jcp.25303] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 01/07/2016] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into bone forming cells. Such ability is compromised in elderly individuals resulting in bone disorders such as osteoporosis, also limiting their clinical usage for cell transplantation and bone tissue engineering strategies. In bone marrow niches, adenine and uracil nucleotides are important local regulators of osteogenic differentiation of MSCs. Nucleotides can be released to the extracellular milieu under both physiological and pathological conditions via (1) membrane cell damage, (2) vesicle exocytosis, (3) ATP-binding cassette transporters, and/or (4) facilitated diffusion through maxi-anion channels, hemichannels or ligand-gated receptor pores. Nucleotides and their derivatives act via adenosine P1 (A1 , A2A , A2B , and A3 ) and nucleotide-sensitive P2 purinoceptors comprising ionotropic P2X and G-protein-coupled P2Y receptors. Purinoceptors activation is terminated by membrane-bound ecto-nucleotidases and other ecto-phosphatases, which rapidly hydrolyse extracellular nucleotides to their respective nucleoside 5'-di- and mono-phosphates, nucleosides and free phosphates, or pyrophosphates. Current knowledge suggests that different players of the "purinome" cascade, namely nucleotide release sites, ecto-nucleotidases and purinoceptors, orchestrate to fine-tuning regulate the activity of MSCs in the bone microenvironment. Increasing studies, using osteoprogenitor cell lines, animal models and, more recently, non-modified MSCs from postmenopausal women, raised the possibility to target chief components of the purinergic signaling pathway to regenerate the ability of aged MSCs to differentiate into functional osteoblasts. This review summarizes the main findings of those studies, prompting for novel therapeutic strategies to control ageing disorders where bone destruction exceeds bone formation, like osteoporosis, rheumatoid arthritis, and fracture mal-union. J. Cell. Physiol. 231: 1852-1861, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- J B Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia-Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar-Universidade do Porto (ICBAS-UP), Portugal
| | - P Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia-Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar-Universidade do Porto (ICBAS-UP), Portugal
| |
Collapse
|
20
|
Abstract
Accumulating evidence now suggests that purinergic signalling exerts significant regulatory effects in the musculoskeletal system. In particular, it has emerged that extracellular nucleotides are key regulators of bone cell differentiation, survival and function. This review discusses our current understanding of the direct effects of purinergic signalling in bone, cartilage and muscle.
Collapse
Affiliation(s)
- Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, United Kingdom.
| |
Collapse
|
21
|
Burnstock G, Arnett TR, Orriss IR. Purinergic signalling in the musculoskeletal system. Purinergic Signal 2013; 9:541-72. [PMID: 23943493 PMCID: PMC3889393 DOI: 10.1007/s11302-013-9381-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 07/12/2013] [Indexed: 12/11/2022] Open
Abstract
It is now widely recognised that extracellular nucleotides, signalling via purinergic receptors, participate in numerous biological processes in most tissues. It has become evident that extracellular nucleotides have significant regulatory effects in the musculoskeletal system. In early development, ATP released from motor nerves along with acetylcholine acts as a cotransmitter in neuromuscular transmission; in mature animals, ATP functions as a neuromodulator. Purinergic receptors expressed by skeletal muscle and satellite cells play important pathophysiological roles in their development or repair. In many cell types, expression of purinergic receptors is often dependent on differentiation. For example, sequential expression of P2X5, P2Y1 and P2X2 receptors occurs during muscle regeneration in the mdx model of muscular dystrophy. In bone and cartilage cells, the functional effects of purinergic signalling appear to be largely negative. ATP stimulates the formation and activation of osteoclasts, the bone-destroying cells. Another role appears to be as a potent local inhibitor of mineralisation. In osteoblasts, the bone-forming cells, ATP acts via P2 receptors to limit bone mineralisation by inhibiting alkaline phosphatase expression and activity. Extracellular ATP additionally exerts significant effects on mineralisation via its hydrolysis product, pyrophosphate. Evidence now suggests that purinergic signalling is potentially important in several bone and joint disorders including osteoporosis, rheumatoid arthritis and cancers. Strategies for future musculoskeletal therapies might involve modulation of purinergic receptor function or of the ecto-nucleotidases responsible for ATP breakdown or ATP transport inhibitors.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK,
| | | | | |
Collapse
|
22
|
Tan TW, Pfau B, Jones D, Meyer T. Stimulation of primary osteoblasts with ATP induces transient vinculin clustering at sites of high intracellular traction force. J Mol Histol 2013; 45:81-9. [PMID: 23933795 PMCID: PMC4544565 DOI: 10.1007/s10735-013-9530-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/25/2013] [Indexed: 01/24/2023]
Abstract
Adenosine 5′-triphosphate (ATP), released in response to mechanical and inflammatory stimuli, induces the dynamic and asynchronous protrusion and subsequent retraction of local membrane structures in osteoblasts. The molecular mechanisms involved in the ligand-stimulated herniation of the plasma membrane are largely unknown, which prompted us to investigate whether the focal-adhesion protein vinculin is engaged in the cytoskeletal alterations that underlie the ATP-induced membrane blebbing. Using time-lapse fluorescence microscopy of primary bovine osteoblast-like cells expressing green fluorescent protein-tagged vinculin, we found that stimulation of cells with 100 μM ATP resulted in the transient and rapid clustering of recombinant vinculin in the cell periphery, starting approximately 100 s after addition of the nucleotide. The ephemeral nature of the vinculin clusters was made evident by the brevity of their mean assembly and disassembly times (66.7 ± 13.3 s and 99.0 ± 6.6 s, respectively). Traction force vector maps demonstrated that the vinculin-rich clusters were localized predominantly at sites of high traction force. Intracellular calcium measurements showed that the ligand-induced increase in [Ca2+]i clearly preceded the clustering of vinculin, since [Ca2+]i levels returned to normal within 30 s of exposure to ATP, indicating that intracellular calcium transients trigger a cascade of signalling events that ultimately result in the incorporation of vinculin into membrane-associated focal aggregates.
Collapse
Affiliation(s)
- Toh Weng Tan
- Institute for Experimental Orthopaedics and Biomechanics, University of Marburg, Marburg, Germany
| | | | | | | |
Collapse
|
23
|
|
24
|
Silber AS, Pfau B, Tan TW, Jacob R, Jones D, Meyer T. Dynamic redistribution of paxillin in bovine osteoblasts stimulated with adenosine 5'-triphosphate. J Mol Histol 2012; 43:571-80. [PMID: 22556032 PMCID: PMC3460167 DOI: 10.1007/s10735-012-9419-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 04/09/2012] [Indexed: 01/21/2023]
Abstract
Exposure to extracellular 5′-adenosine triphosphate (ATP) is known to induce membrane blebbing. In this study, we investigated the subcellular distribution of the cytoskeletal adaptor protein paxillin in primary bovine osteoblasts upon stimulation with ATP. Cells expressing a fusion protein of green fluorescent protein (GFP) and paxillin were followed by time-lapse video-microscopy after stimulation with 100 μM ATP. Within 100 s, GFP-paxillin became incorporated in numerous de novo formed focal aggregates localized at the cell periphery. The assembly of individual paxillin-containing aggregates occurred with a mean half-life time of <60 s, whereas their disassembly lasted twice as long. Despite the ongoing presence of ATP, the formation of paxillin aggregates was self-limiting within 25 min. Paxillin clustering was preceded by a transient rise in cytoplasmic calcium transients, which peaked already 20 s after adding ATP. The high mobility of paxillin was confirmed by measuring the dissociation rate of GFP-paxillin at mature focal adhesions, demonstrating the presence of a highly mobile fraction with a mean recovery half-life of 8.2 ± 1.2 s, followed by a slower phase (53 ± 20 s). Thus, both the exchange of paxillin at mature focal adhesions and the increase in intracellular calcium concentrations upon ATP stimulation are very rapid processes, which override the time course of ATP-induced paxillin membrane clustering by one to two orders of magnitude. Our data demonstrate that the transient recruitment of paxillin in membrane protuberances is based on the high intracytoplasmic mobility of unbound paxillin molecules and their rapid focal accumulation.
Collapse
Affiliation(s)
- Ann-Sophie Silber
- Institut für Experimentelle Orthopädie und Biomechanik, Philipps-Universität Marburg, Marburg, Germany
| | | | | | | | | | | |
Collapse
|
25
|
Interaction of purinergic receptors with GPCRs, ion channels, tyrosine kinase and steroid hormone receptors orchestrates cell function. Purinergic Signal 2011; 8:91-103. [PMID: 21887492 DOI: 10.1007/s11302-011-9260-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 08/16/2011] [Indexed: 01/26/2023] Open
Abstract
Extracellular purines and pyrimidines have emerged as key regulators of a wide range of physiological and pathophysiological cellular processes acting through P1 and P2 cell surface receptors. Increasing evidence suggests that purinergic receptors can interact with and/or modulate the activity of other classes of receptors and ion channels. This review will focus on the interactions of purinergic receptors with other GPCRs, ion channels, receptor tyrosine kinases, and steroid hormone receptors. Also, the signal transduction pathways regulated by these complexes and their new functional properties are discussed.
Collapse
|
26
|
Activation of the PI3K/Akt signaling pathway through P2Y₂ receptors by extracellular ATP is involved in osteoblastic cell proliferation. Arch Biochem Biophys 2011; 513:144-52. [PMID: 21763267 DOI: 10.1016/j.abb.2011.06.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 06/28/2011] [Accepted: 06/29/2011] [Indexed: 12/31/2022]
Abstract
We studied the PI3K/Akt signaling pathway modulation and its involvement in the stimulation of ROS 17/2.8 osteoblast-like cell proliferation by extracellular ATP. A dose- and time-dependent increase in Akt-Ser 473 phosphorylation (p-Akt) was observed. p-Akt was increased by ATPγS and UTP, but not by ADPβS. Akt activation was abolished by PI3K inhibitors and reduced by inhibitors of PI-PLC, Src, calmodulin (CaM) but not of CaMK. p-Akt was diminished by cell incubation in a Ca²⁺-free medium but not by the use of L-type calcium channel blockers. The rise in intracellular Ca²⁺ induced by ATP was potentiated in the presence of Ro318220, a PKC inhibitor, and attenuated by the TPA, a known activator of PKC. ATP-dependent p-Akt was diminished by TPA and augmented by Ro318220 treatment in a Ca²⁺-containing but not in a Ca²⁺-free medium. ATP stimulated the proliferation of both ROS 17/2.8 cells and rat osteoblasts through PI3K/Akt. In the primary osteoblasts, ATP induces alkaline phosphatase activity via PI3K, suggesting that the nucleotide promotes osteoblast differentiation. These results suggest that ATP stimulates osteoblast proliferation through PI-PLC linked-P2Y₂ receptors and PI3K/Akt pathway activation involving Ca²⁺, CaM and Src. PKC seems to regulate Akt activation through Src and the Ca²⁺ influx/CaM pathway.
Collapse
|
27
|
Protein phosphatases: possible bisphosphonate binding sites mediating stimulation of osteoblast proliferation. Arch Biochem Biophys 2010; 507:248-53. [PMID: 21167123 DOI: 10.1016/j.abb.2010.12.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 12/09/2010] [Accepted: 12/10/2010] [Indexed: 11/24/2022]
Abstract
We investigated the existence of a bisphosphonate (BP) target site in osteoblasts. Binding assays using [³H]-olpadronate ([³H]OPD) in whole cells showed the presence of specific, saturable and high affinity binding for OPD (K(d)=1.39 ± 0.33 μM) in osteoblasts. [³H]OPD was displaced from its binding site by micromolar concentrations of lidadronate, alendronate and etidronate (K(d)=1.42 ± 0.15 μM, 2.00 ± 0.2 μM and 2.4 ± 0.4 μM, respectively), and by millimolar concentrations of the non-permeant protein phosphatase (PP) substrates p-nitrophenylphosphate and α-naphtylphosphate. PP inhibitors orthovanadate, NaF or vpb(bipy) did not displace [³H]OPD. As expected, specific OPD binding was detected in the plasma membrane of ROS 17/2.8 cells, although significant BP binding was also found intracellularly. Moreover, OPD increased DNA synthesis in these cells with a temporal profile similar to the protein tyrosine phosphatase (PTP) inhibitors, Na₃VO₄ and vpb(bipy); but different from a general PP inhibitor (NaF). The stimulatory effect of OPD and PTP inhibitors on osteoblast proliferation was inhibited by the protein tyrosine kinase inhibitors genistein and geldanamycin. These results provide new evidence on the existence of a BP target in osteoblastic cells, presumably a PTP, which may be involved in the stimulatory action of BPs on osteoblast proliferation.
Collapse
|
28
|
Muscarinic acetylcholine receptors present in human osteoblast and bone tissue. Eur J Pharmacol 2010; 650:34-40. [PMID: 20888332 DOI: 10.1016/j.ejphar.2010.09.031] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 07/29/2010] [Accepted: 09/14/2010] [Indexed: 01/27/2023]
Abstract
Acetylcholine is the predominant neurotransmitter in the neuromuscular junction, and a role in bone has been postulated. The expression of nicotinic receptors has been reported in osteoblasts, but the expression and function of muscarinic receptor in bone remain obscure. In this study, we investigated the expression and functional activities of muscarinic receptor subtypes in human osteoblast cell lines and animal and human bone tissue. The mRNA levels of muscarinic receptor subtypes were detected by reverse-transcription polymerase chain reaction. We found that muscarinic subtypes m1, m2, m3, m4, and m5 were expressed at different levels in human osteosarcoma HOS cells, rat femur, and human rib bone tissue; m1, m4, m5 were in cultured mouse femur bone cells and cultured mouse calvarial bone cells; m2, m3, m4 were in bovine bone. The mRNA of neuronal markers, light-, medium- and heavy-neurofilament, was not found in human bone tissues to exclude the possible contamination from neuronal tissue. Methacholine induced an elevation in cytosolic calcium concentration and proliferation in HOS cells. Both effects were blocked by atropine. We conclude that muscarinic receptor is present in bone tissue to evoke calcium signaling and modulate cell proliferation. Different muscarinic receptor subtypes are distributed in various parts of the animal skeletal system including the different species and bone portions. Bone remodeling involving osteoblast proliferation leads the possibilities that muscarinic receptor may play roles in bone remodeling.
Collapse
|
29
|
Bilbao PS, Santillán G, Boland R. ATP stimulates the proliferation of MCF-7 cells through the PI3K/Akt signaling pathway. Arch Biochem Biophys 2010; 499:40-8. [PMID: 20450878 DOI: 10.1016/j.abb.2010.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 04/28/2010] [Accepted: 05/01/2010] [Indexed: 11/30/2022]
Abstract
We studied the modulation of the PI3K/Akt signaling pathway by ATP in MCF-7 cells. Western blot analysis showed that ATP stimulated the phosphorylation of Akt in a dose- and time-dependent manner. Akt phosphorylation in response to nucleotides followed the potency order ATP=UTP=ATPgammaS>>ADP=UDP>ADPbetaS=adenosine, suggesting participation of P2Y(2/4) receptors. Inhibitors of PI3K, PLC, PKC and Src or Src antisense oligonucleotides prevented ATP-induced phosphorylation of Akt. Incubation of cells with 2-APB or in a nominally Ca(2+)-free medium plus EGTA showed that Akt phosphorylation by ATP depends on intracellular calcium release but is independent of calcium influx. The PI3K inhibitor was not effective in reducing MAPKs phosphorylation by ATP. ATP and UTP stimulated MCF-7 cell proliferation, effect that was inhibited by PI3K, PLC, PKC, Src and MAPKs inhibitors. These findings suggest that ATP modulation of P2Y(2/4) receptors increases MCF-7 cell proliferation by activation of the PI3K/Akt signaling pathway through PLC/IP(3)/Ca(2+), PKC and Src.
Collapse
Affiliation(s)
- Paola Scodelaro Bilbao
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, San Juan 670, (B8000ICN) Bahía Blanca, Argentina
| | | | | |
Collapse
|
30
|
ATP modulates transcription factors through P2Y2 and P2Y4 receptors via PKC/MAPKs and PKC/Src pathways in MCF-7 cells. Arch Biochem Biophys 2009; 494:7-14. [PMID: 19900397 DOI: 10.1016/j.abb.2009.11.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 11/03/2009] [Accepted: 11/04/2009] [Indexed: 02/04/2023]
Abstract
In this work, we studied the involvement of PKC and Src in the phosphorylation of ERK1/2, p38 and JNK1 MAPKs and in the modulation of ATF-1, c-Fos, c-Jun and Jun D transcription factors by ATP in MCF-7 breast cancer cells. RT-PCR studies and nucleotide sequence analysis confirmed first the expression of P2Y(2)- and P2Y(4)-receptor subtypes. The use of specific inhibitors and Src antisense oligonucleotides showed that PKC, but not Src, plays a role in the phosphorylation of MAPKs by ATP. ATP stimulated the expression of c-Fos and the phosphorylation c-Jun, Jun D and ATF-1. PKC and Src only participated in c-Fos induction and in ATF-1 phosphorylation. Pharmacological inhibition of MAPKs demonstrated that c-Fos induction and phosphorylation of c-Jun and Jun D, but not of ATF-1, depend on MAPK activation. These results suggest that stimulation of P2Y(2) and P2Y(4) receptors by ATP modulates transcription factors through PKC/MAPKs and PKC/Src pathways in MCF-7 cells.
Collapse
|
31
|
Buzzi N, Bilbao PS, Boland R, de Boland AR. Extracellular ATP activates MAP kinase cascades through a P2Y purinergic receptor in the human intestinal Caco-2 cell line. Biochim Biophys Acta Gen Subj 2009; 1790:1651-9. [PMID: 19836435 DOI: 10.1016/j.bbagen.2009.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 09/14/2009] [Accepted: 10/08/2009] [Indexed: 11/26/2022]
Abstract
BACKGROUND ATP exerts diverse effects on various cell types via specific purinergic P2Y receptors. Intracellular signaling cascades are the main routes of communication between P2Y receptors and regulatory targets in the cell. METHODS AND RESULTS We examined the role of ATP in the modulation of ERK1/2, JNK1/2, and p38 MAP kinases (MAPKs) in human colon cancer Caco-2 cells. Immunoblot analysis showed that ATP induces the phosphorylation of MAPKs in a time- and dose-dependent manner, peaking at 5 min at 10 microM ATP. Moreover, ATPgammaS, UTP, and UDP but not ADP or ADPbetaS increased phosphorylation of MAPKs, indicating the involvement of, at least, P2Y2/P2Y4 and P2Y6 receptor subtypes. RT-PCR studies and PCR product sequencing supported the expression of P2Y2 and P2Y4 receptors in this cell line. Spectrofluorimetric measurements showed that cell stimulation with ATP induced transient elevations in intracellular calcium concentration. In addition, ATP-induced phosphorylation of MAPKs in Caco-2 cells was dependent on Src family tyrosine kinases, calcium influx, and intracellular Ca2+ release and was partially dependent on the cAMP/PKA and PKC pathways and the EGFR. GENERAL SIGNIFICANCE These findings provide new molecular basis for further understanding the mechanisms involved in ATP functions, as a signal transducer and activator of MAP kinase cascades, in colon adenocarcinoma Caco-2 cells.
Collapse
Affiliation(s)
- Natalia Buzzi
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, San Juan 670, 8000 Bahía Blanca, Argentina
| | | | | | | |
Collapse
|