1
|
Wakefield B, Tang J, Hutchinson JL, Kanji R, Brooks C, Grol MW, Séguin CA, Penuela S, Beier F. Pannexin 3 deletion in mice results in knee osteoarthritis and intervertebral disc degeneration after forced treadmill running. J Orthop Res 2024; 42:1696-1709. [PMID: 38499500 DOI: 10.1002/jor.25830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/10/2023] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
Pannexin 3 (Panx3) is a glycoprotein that forms mechanosensitive channels expressed in chondrocytes and annulus fibrosus cells of the intervertebral disc (IVD). Evidence suggests Panx3 plays contrasting roles in traumatic versus aging osteoarthritis (OA) and intervertebral disc degeneration (IDD). However, whether its deletion influences the response of joint tissue to forced use is unknown. The purpose of this study was to determine if Panx3 deletion in mice causes increased knee joint OA and IDD after forced treadmill running. Male and female wildtype (WT) and Panx3 knockout (KO) mice were randomized to either a no-exercise group (sedentary; SED) or daily forced treadmill running (forced exercise; FEX) from 24 to 30 weeks of age. Knee cartilage and IVD histopathology were evaluated by histology, while tibial secondary ossification centers were analyzed using microcomputed tomography (µCT). Both male and female Panx3 KO mice developed larger superficial defects of the tibial cartilage after forced treadmill running compared with SED WT mice. Additionally, Panx3 KO mice developed reduced bone volume, and female PANX3 KO mice had lengthening of the lateral tubercle at the intercondylar eminence. In the lower lumbar spine, both male and female Panx3 KO mice developed histopathological features of IDD after running compared to SED WT mice. These findings suggest that the combination of deleting Panx3 and forced treadmill running induces OA and causes histopathological changes associated with the degeneration of the IVDs in mice.
Collapse
Affiliation(s)
- Brent Wakefield
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
| | - Justin Tang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
| | - Jeffrey L Hutchinson
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Rehanna Kanji
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Courtney Brooks
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Matthew W Grol
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Cheryle A Séguin
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
| | - Frank Beier
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
2
|
Luo Y, Zheng S, Xiao W, Zhang H, Li Y. Pannexins in the musculoskeletal system: new targets for development and disease progression. Bone Res 2024; 12:26. [PMID: 38705887 PMCID: PMC11070431 DOI: 10.1038/s41413-024-00334-8] [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: 12/08/2023] [Revised: 03/04/2024] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
During cell differentiation, growth, and development, cells can respond to extracellular stimuli through communication channels. Pannexin (Panx) family and connexin (Cx) family are two important types of channel-forming proteins. Panx family contains three members (Panx1-3) and is expressed widely in bone, cartilage and muscle. Although there is no sequence homology between Panx family and Cx family, they exhibit similar configurations and functions. Similar to Cxs, the key roles of Panxs in the maintenance of physiological functions of the musculoskeletal system and disease progression were gradually revealed later. Here, we seek to elucidate the structure of Panxs and their roles in regulating processes such as osteogenesis, chondrogenesis, and muscle growth. We also focus on the comparison between Cx and Panx. As a new key target, Panxs expression imbalance and dysfunction in muscle and the therapeutic potentials of Panxs in joint diseases are also discussed.
Collapse
Affiliation(s)
- Yan Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, 410008, China
| | - Shengyuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, 410008, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hang Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
3
|
O'Donnell BL, Penuela S. Skin in the game: pannexin channels in healthy and cancerous skin. Biochem J 2023; 480:1929-1949. [PMID: 38038973 DOI: 10.1042/bcj20230176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
The skin is a highly organized tissue composed of multiple layers and cell types that require coordinated cell to cell communication to maintain tissue homeostasis. In skin cancer, this organized structure and communication is disrupted, prompting the malignant transformation of healthy cells into melanoma, basal cell carcinoma or squamous cell carcinoma tumours. One such family of channel proteins critical for cellular communication is pannexins (PANX1, PANX2, PANX3), all of which are present in the skin. These heptameric single-membrane channels act as conduits for small molecules and ions like ATP and Ca2+ but have also been shown to have channel-independent functions through their interacting partners or action in signalling pathways. Pannexins have diverse roles in the skin such as in skin development, aging, barrier function, keratinocyte differentiation, inflammation, and wound healing, which were discovered through work with pannexin knockout mice, organotypic epidermis models, primary cells, and immortalized cell lines. In the context of cutaneous cancer, PANX1 is present at high levels in melanoma tumours and functions in melanoma carcinogenesis, and both PANX1 and PANX3 expression is altered in non-melanoma skin cancer. PANX2 has thus far not been implicated in any skin cancer. This review will discuss pannexin isoforms, structure, trafficking, post-translational modifications, interactome, and channel activity. We will also outline the expression, localization, and function of pannexin channels within the diverse cell types of the epidermis, dermis, hypodermis, and adnexal structures of the skin, and how these properties are exploited or abrogated in instances of skin cancer.
Collapse
Affiliation(s)
- Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
- Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| |
Collapse
|
4
|
Wu YL, Yang AH, Chiu YH. Recent advances in the structure and activation mechanisms of metabolite-releasing Pannexin 1 channels. Biochem Soc Trans 2023; 51:1687-1699. [PMID: 37622532 DOI: 10.1042/bst20230038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
Pannexin 1 (PANX1) is a widely expressed large-pore ion channel located in the plasma membrane of almost all vertebrate cells. It possesses a unique ability to act as a conduit for both inorganic ions (e.g. potassium or chloride) and bioactive metabolites (e.g. ATP or glutamate), thereby activating varying signaling pathways in an autocrine or paracrine manner. Given its crucial role in cell-cell interactions, the activity of PANX1 has been implicated in maintaining homeostasis of cardiovascular, immune, and nervous systems. Dysregulation of PANX1 has also been linked to numerous diseases, such as ischemic stroke, seizure, and inflammatory disorders. Therefore, the mechanisms underlying different modes of PANX1 activation and its context-specific channel properties have gathered significant attention. In this review, we summarize the roles of PANX1 in various physiological processes and diseases, and analyze the accumulated lines of evidence supporting diverse molecular mechanisms associated with different PANX1 activation modalities. We focus on examining recent discoveries regarding PANX1 regulations by reversible post-translational modifications, elevated intracellular calcium concentration, and protein-protein interactions, as well as by irreversible cleavage of its C-terminal tail. Additionally, we delve into the caveats in the proposed PANX1 gating mechanisms and channel open-closed configurations by critically analyzing the structural insights derived from cryo-EM studies and the unitary properties of PANX1 channels. By doing so, we aim to identify potential research directions for a better understanding of the functions and regulations of PANX1 channels.
Collapse
Affiliation(s)
- Yi-Ling Wu
- Department of Life Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
| | - Ai-Hsing Yang
- Department of Life Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
| | - Yu-Hsin Chiu
- Department of Life Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, No. 101, Sec. 2, Kuang Fu Rd., Hsinchu 300044, Taiwan
| |
Collapse
|
5
|
Atf7ip Inhibits Osteoblast Differentiation via Negative Regulation of the Sp7 Transcription Factor. Int J Mol Sci 2023; 24:ijms24054305. [PMID: 36901736 PMCID: PMC10002255 DOI: 10.3390/ijms24054305] [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: 01/02/2023] [Revised: 02/03/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Epigenetic modifications are critical for cell differentiation and growth. As a regulator of H3K9 methylation, Setdb1 is implicated in osteoblast proliferation and differentiation. The activity and nucleus localization of Setdb1 are regulated by its binding partner, Atf7ip. However, whether Atf7ip is involved in the regulation of osteoblast differentiation remains largely unclear. In the present study, we found that Atf7ip expression was upregulated during the osteogenesis of primary bone marrow stromal cells and MC3T3-E1 cells, and was induced in PTH-treated cells. The overexpression of Atf7ip impaired osteoblast differentiation in MC3T3-E1 cells regardless of PTH treatment, as measured by the expression of osteoblast differentiation markers, Alp-positive cells, Alp activity, and calcium deposition. Conversely, the depletion of Atf7ip in MC3T3-E1 cells promoted osteoblast differentiation. Compared with the control mice, animals with Atf7ip deletion in the osteoblasts (Oc-Cre;Atf7ipf/f) showed more bone formation and a significant increase in the bone trabeculae microarchitecture, as reflected by μ-CT and bone histomorphometry. Mechanistically, Atf7ip contributed to the nucleus localization of Setdb1 in MC3T3-E1, but did not affect Setdb1 expression. Atf7ip negatively regulated Sp7 expression, and through specific siRNA, Sp7 knockdown attenuated the enhancing role of Atf7ip deletion in osteoblast differentiation. Through these data, we identified Atf7ip as a novel negative regulator of osteogenesis, possibly via its epigenetic regulation of Sp7 expression, and demonstrated that Atf7ip inhibition is a potential therapeutic measure for enhancing bone formation.
Collapse
|
6
|
Jiang P, Ma X, Han S, Ma L, Ai J, Wu L, Zhang Y, Xiao H, Tian M, Tao WA, Zhang S, Chai R. Characterization of the microRNA transcriptomes and proteomics of cochlear tissue-derived small extracellular vesicles from mice of different ages after birth. Cell Mol Life Sci 2022; 79:154. [PMID: 35218422 PMCID: PMC11072265 DOI: 10.1007/s00018-022-04164-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/30/2021] [Accepted: 01/23/2022] [Indexed: 12/22/2022]
Abstract
The cochlea is an important sensory organ for both balance and sound perception, and the formation of the cochlea is a complex developmental process. The development of the mouse cochlea begins on embryonic day (E)9 and continues until postnatal day (P)21 when the hearing system is considered mature. Small extracellular vesicles (sEVs), with a diameter ranging from 30 to 200 nm, have been considered a significant medium for information communication in both physiological and pathological processes. However, there are no studies exploring the role of sEVs in the development of the cochlea. Here, we isolated tissue-derived sEVs from the cochleae of FVB mice at P3, P7, P14, and P21 by ultracentrifugation. These sEVs were first characterized by transmission electron microscopy, nanoparticle tracking analysis, and western blotting. Next, we used small RNA-seq and mass spectrometry to characterize the microRNA transcriptomes and proteomes of cochlear sEVs from mice at different ages. Many microRNAs and proteins were discovered to be related to inner ear development, anatomical structure development, and auditory nervous system development. These results all suggest that sEVs exist in the cochlea and are likely to be essential for the normal development of the auditory system. Our findings provide many sEV microRNA and protein targets for future studies of the roles of cochlear sEVs.
Collapse
Affiliation(s)
- Pei Jiang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xiangyu Ma
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Shanying Han
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Leyao Ma
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jingru Ai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Leilei Wu
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yuan Zhang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Hairong Xiao
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Mengyao Tian
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - W Andy Tao
- Department of Chemistry, Department of Biochemistry, Purdue University, West Lafayette, Indiana, 47907, USA.
- Center for Cancer Research, Purdue University, West Lafayette, Indiana, 47907, USA.
| | - Shasha Zhang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
7
|
Larrañaga-Vera A, Marco-Bonilla M, Largo R, Herrero-Beaumont G, Mediero A, Cronstein B. ATP transporters in the joints. Purinergic Signal 2021; 17:591-605. [PMID: 34392490 PMCID: PMC8677878 DOI: 10.1007/s11302-021-09810-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023] Open
Abstract
Extracellular adenosine triphosphate (ATP) plays a central role in a wide variety of joint diseases. ATP is generated intracellularly, and the concentration of the extracellular ATP pool is determined by the regulation of its transport out of the cell. A variety of ATP transporters have been described, with connexins and pannexins the most commonly cited. Both form intercellular channels, known as gap junctions, that facilitate the transport of various small molecules between cells and mediate cell-cell communication. Connexins and pannexins also form pores, or hemichannels, that are permeable to certain molecules, including ATP. All joint tissues express one or more connexins and pannexins, and their expression is altered in some pathological conditions, such as osteoarthritis (OA) and rheumatoid arthritis (RA), indicating that they may be involved in the onset and progression of these pathologies. The aging of the global population, along with increases in the prevalence of obesity and metabolic dysfunction, is associated with a rising frequency of joint diseases along with the increased costs and burden of related illness. The modulation of connexins and pannexins represents an attractive therapeutic target in joint disease, but their complex regulation, their combination of gap-junction-dependent and -independent functions, and their interplay between gap junction and hemichannel formation are not yet fully elucidated. In this review, we try to shed light on the regulation of these proteins and their roles in ATP transport to the extracellular space in the context of joint disease, and specifically OA and RA.
Collapse
Affiliation(s)
- Ane Larrañaga-Vera
- Department of Medicine, Division of Translational Medicine, NYU Langone Health, New York, NY, USA
| | - Miguel Marco-Bonilla
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, 28040, Madrid, Spain
| | - Raquel Largo
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, 28040, Madrid, Spain
| | | | - Aránzazu Mediero
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, 28040, Madrid, Spain.
| | - Bruce Cronstein
- Department of Medicine, Division of Translational Medicine, NYU Langone Health, New York, NY, USA
| |
Collapse
|
8
|
Laird DW, Penuela S. Pannexin biology and emerging linkages to cancer. Trends Cancer 2021; 7:1119-1131. [PMID: 34389277 DOI: 10.1016/j.trecan.2021.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/18/2022]
Abstract
Pannexins are a family of glycoproteins that comprises three members, PANX1, PANX2, and PANX3. The widely expressed and interrogated PANX1 forms heptameric membrane channels that primarily serve to connect the cytoplasm to the extracellular milieu by being selectively permeable to small signaling molecules when activated. Apart from notable exceptions, PANX1 in many tumor cells appears to facilitate tumor growth and metastasis, suggesting that pannexin-blocking therapeutics may have utility in cancer. Attenuation of PANX1 function must also consider the fact that PANX1 is found in stromal cells of the tumor microenvironment (TME), including immune cells. This review highlights the key discoveries of the past 5 years that suggest pannexins facilitate, or in some cases inhibit, tumor cell growth and metastasis via direct protein interactions and through the regulated efflux of signaling molecules.
Collapse
Affiliation(s)
- Dale W Laird
- Department of Anatomy and Cell Biology, and Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Department of Oncology, Divisions of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
9
|
O'Donnell BL, Penuela S. Pannexin 3 channels in health and disease. Purinergic Signal 2021; 17:577-589. [PMID: 34250568 DOI: 10.1007/s11302-021-09805-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/30/2021] [Indexed: 01/07/2023] Open
Abstract
Pannexin 3 (PANX3) is a member of the pannexin family of single membrane channel-forming glycoproteins. Originally thought to have a limited localization in cartilage, bone, and skin, PANX3 has now been detected in a variety of other tissues including skeletal muscle, mammary glands, the male reproductive tract, the cochlea, blood vessels, small intestines, teeth, and the vomeronasal organ. In many cell types of the musculoskeletal system, such as osteoblasts, chondrocytes, and odontoblasts, PANX3 has been shown to regulate the balance of proliferation and differentiation. PANX3 can be induced during progenitor cell differentiation, functioning at the cell surface as a conduit for ATP and/or in the endoplasmic reticulum as a calcium leak channel. Evidence in osteoblasts and monocytes also highlight a role for PANX3 in purinergic signalling through its function as an ATP release channel. PANX3 is critical in the development and ageing of bone and cartilage, with its levels temporally regulated in other tissues such as skeletal muscle, skin, and the cochlea. In diseases such as osteoarthritis and intervertebral disc degeneration, PANX3 can have either protective or detrimental roles depending on if the disease is age-related or injury-induced. This review will discuss PANX3 function in tissue growth and regeneration, its role in cellular differentiation, and how it becomes dysregulated in disease conditions such as obesity, Duchenne's muscular dystrophy, osteosarcoma, and non-melanoma skin cancer, where most of the findings on PANX3 function can be attributed to the characterization of Panx3 KO mouse models.
Collapse
Affiliation(s)
- Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
- Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
| |
Collapse
|
10
|
Noort RJ, Christopher GA, Esseltine JL. Pannexin 1 Influences Lineage Specification of Human iPSCs. Front Cell Dev Biol 2021; 9:659397. [PMID: 33937260 PMCID: PMC8086557 DOI: 10.3389/fcell.2021.659397] [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] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Every single cell in the body communicates with nearby cells to locally organize activities with their neighbors and dysfunctional cell-cell communication can be detrimental during cell lineage commitment, tissue patterning and organ development. Pannexin channels (PANX1, PANX2, and PANX3) facilitate purinergic paracrine signaling through the passage of messenger molecules out of cells. PANX1 is widely expressed throughout the body and has recently been identified in human oocytes as well as 2 and 4-cell stage human embryos. Given its abundance across multiple adult tissues and its expression at the earliest stages of human development, we sought to understand whether PANX1 impacts human induced pluripotent stem cells (iPSCs) or plays a role in cell fate decisions. Western blot, immunofluorescence and flow cytometry reveal that PANX1 is expressed in iPSCs as well as all three germ lineages derived from these cells: ectoderm, endoderm, and mesoderm. PANX1 demonstrates differential glycosylation patterns and subcellular localization across the germ lineages. Using CRISPR-Cas9 gene ablation, we find that loss of PANX1 has no obvious impact on iPSC morphology, survival, or pluripotency gene expression. However, PANX1 gene knockout iPSCs exhibit apparent lineage specification bias under 3-dimensional spontaneous differentiation into the three germ lineages. Indeed, loss of PANX1 increases representation of endodermal and mesodermal populations in PANX1 knockout cells. Importantly, PANX1 knockout iPSCs are fully capable of differentiating toward each specific lineage when exposed to the appropriate external signaling pressures, suggesting that although PANX1 influences germ lineage specification, it is not essential to this process.
Collapse
Affiliation(s)
- Rebecca J Noort
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Grace A Christopher
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jessica L Esseltine
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Novel therapies for damaged and diseased bone are being developed in a preclinical testing process consisting of in vitro cell experiments followed by in vivo animal studies. The in vitro results are often not representative of the results observed in vivo. This could be caused by the complexity of the natural bone environment that is missing in vitro. Ex vivo bone explant cultures provide a model in which cells are preserved in their native three-dimensional environment. Herein, it is aimed to review the current status of bone explant culture models in relation to their potential in complementing the preclinical evaluation process with specific attention paid to the incorporation of mechanical loading within ex vivo culture systems. RECENT FINDINGS Bone explant cultures are often performed with physiologically less relevant bone, immature bone, and explants derived from rodents, which complicates translatability into clinical practice. Mature bone explants encounter difficulties with maintaining viability, especially in static culture. The integration of mechanical stimuli was able to extend the lifespan of explants and to induce new bone formation. Bone explant cultures provide unique platforms for bone research and mechanical loading was demonstrated to be an important component in achieving osteogenesis ex vivo. However, more research is needed to establish a representative, reliable, and reproducible bone explant culture system that includes both components of bone remodeling, i.e., formation and resorption, in order to bridge the gap between in vitro and in vivo research in preclinical testing.
Collapse
Affiliation(s)
- E E A Cramer
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - K Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - S Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands.
| |
Collapse
|
12
|
Komatsu DE, Duque E, Hadjiargyrou M. MicroRNAs and fracture healing: Pre-clinical studies. Bone 2021; 143:115758. [PMID: 33212318 PMCID: PMC7769985 DOI: 10.1016/j.bone.2020.115758] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/28/2022]
Abstract
During the past several years, pre-clinical experiments have established that microRNAs (miRNAs), small non-coding RNAs, serve as key regulatory molecules of fracture healing. Their easy modulation with agonists and antagonists make them highly desirable targets for future therapeutic strategies, especially for pathophysiologic fractures that either do not heal (nonunions) or are delayed. It is now well documented that these problematic fractures lead to human suffering and impairment of life quality. Additionally, financial difficulties are also encountered as work productivity decreases and income is reduced. Moreover, targeting miRNAs may also be an avenue to enhancing normal physiological fracture healing. Herein we present the most current knowledge of the involvement of miRNAs during fracture healing in pre-clinical studies. Following a brief description on the nature of miRNAs and of the fracture healing process, we present data from studies focusing specifically, on miRNA regulation of osteoblast differentiation and osteogenesis (within the context of known signaling pathways), chondrocytes, angiogenesis, and apoptosis, all critical to successful bone repair. Further, we also discuss miRNAs and exosomes. We hope that this manuscript serves as a comprehensive review that will facilitate basic/translational scientists in the orthopaedic arena to realize and further decipher the biological and future therapeutic impact of these small regulatory RNA molecules, especially as they relate to the molecular events of each of the major phases of fracture healing.
Collapse
Affiliation(s)
- David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Edie Duque
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, United States of America.
| |
Collapse
|
13
|
Zhang P, Ishikawa M, Doyle A, Nakamura T, He B, Yamada Y. Pannexin 3 regulates skin development via Epiprofin. Sci Rep 2021; 11:1779. [PMID: 33469169 PMCID: PMC7815752 DOI: 10.1038/s41598-021-81074-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 12/28/2020] [Indexed: 12/11/2022] Open
Abstract
Pannexin 3 (Panx3), a member of the gap junction pannexin family is required for the development of hard tissues including bone, cartilage and teeth. However, the role of Panx3 in skin development remains unclear. Here, we demonstrate that Panx3 regulates skin development by modulating the transcription factor, Epiprofin (Epfn). Panx3-/- mice have impaired skin development and delayed hair follicle regeneration. Loss of Panx3 in knockout mice and suppression by shRNA both elicited a reduction of Epfn expression in the epidermis. In cell culture, Panx3 overexpression promoted HaCaT cell differentiation, cell cycle exit and enhanced Epfn expression. Epfn-/- mice and inhibition of Epfn by siRNA showed no obvious differences of Panx3 expression. Furthermore, Panx3 promotes Akt/NFAT signaling pathway in keratinocyte differentiation by both Panx3 ATP releasing channel and ER Ca2+ channel functions. Our results reveal that Panx3 has a key role factor for the skin development by regulating Epfn.
Collapse
Affiliation(s)
- Peipei Zhang
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Masaki Ishikawa
- Division of Operative Dentistry, Department of Restorative Dentistry, Tohoku University, Graduate School of Dentistry 4-1, Seiryo chou, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Andrew Doyle
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Takashi Nakamura
- Division of Molecular Pharmacology and Cell Biophysics, Department of Oral Biology, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan
| | - Bing He
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yoshihiko Yamada
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
14
|
Ishikawa M, Williams G, Forcinito P, Ishikawa M, Petrie RJ, Saito K, Fukumoto S, Yamada Y. Pannexin 3 ER Ca 2+ channel gating is regulated by phosphorylation at the Serine 68 residue in osteoblast differentiation. Sci Rep 2019; 9:18759. [PMID: 31822768 PMCID: PMC6904572 DOI: 10.1038/s41598-019-55371-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/19/2019] [Indexed: 12/31/2022] Open
Abstract
Pannexin 3 (Panx3) is a regulator of bone formation. Panx3 forms three distinct functional channels: hemichannels, gap junctions, and endoplasmic reticulum (ER) Ca2+ channels. However, the gating mechanisms of the Panx3 channels remain unclear. Here, we show that the Panx3 ER Ca2+ channel is modulated by phosphorylation of the serine 68 residue (Ser68) to promote osteoblast differentiation. Among the 17 candidate phosphorylation sites identified, the mutation of Ser68 to Ala (Ser68Ala) was sufficient to inhibit Panx3-mediated osteoblast differentiation via reduction of Osterix and ALP expression. Using a Ser68 phospho-specific antibody (P-Panx3) revealed Panx3 was phosphorylated in prehypertrophic, hypertrophic chondrocytes, and bone areas of the newborn growth plate. In osteogenic C2C12 cells, P-Panx3 was located on the ER membranes. Importantly, the Ser68Ala mutation only affected Panx3 ER Ca2+ channel function. Ser68 on Panx3 was phosphorylated by ATP stimulation and PI3K/Akt signaling. Finally, real-time FRET imaging and ratio analysis revealed that the Panx3 channel conformation was sensitive to ATP. Together, the phosphorylation of Panx3 at Ser68 is an essential step controlling the gating of the Panx3 ER Ca2+ channel to promote osteogenesis.
Collapse
Affiliation(s)
- Masaki Ishikawa
- Operative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan.
| | - Geneva Williams
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Philadelphia, USA
| | - Patricia Forcinito
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Philadelphia, USA
| | - Momoko Ishikawa
- Department of Pediatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai, 980-8576, Japan
| | - Ryan J Petrie
- Department of Biology, Drexel University, Philadelphia, PA, 19104, USA
| | - Kan Saito
- Department of Pediatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai, 980-8576, Japan
| | - Satoshi Fukumoto
- Department of Pediatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai, 980-8576, Japan
| | - Yoshihiko Yamada
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Philadelphia, USA
| |
Collapse
|
15
|
Jia HL, Zhou DS. Retracted: Downregulation of microRNA-367 promotes osteoblasts growth and proliferation of mice during fracture by activating the PANX3-mediated Wnt/β-catenin pathway. J Cell Biochem 2019; 120:8247-8258. [PMID: 30556206 DOI: 10.1002/jcb.28108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/29/2018] [Indexed: 02/02/2023]
Abstract
A majority of people suffering from bone fractures fail to heal and develop a nonunion, which is a challenging orthopedic complication requiring complex and expensive treatment. Previous data showed the inhibition of some microRNAs (miRNAs or miRs) can enhance fracture healing. The objective of the present study is to explore effects of miR-367 on the osteoblasts growth and proliferation of mouse during fracture via the Wnt/β-catenin pathway by targeting PANX3. Primarily, the femur fracture model was successfully established in 66 (C57BL/6) 6-week-old male mice. To verify whether miR-367 target PANX3, we used the target prediction program and performed luciferase activity determination. Subsequently, to figure out the underlying regulatory roles of miR-367 in fracture, osteoblasts were elucidated by treatment with miR-367 mimic, miR-367 inhibitor, or siRNA against PANX3 to determine the expression of miR-367, siPANX3, β-catenin, and Wnt5b as well as cell proliferation and apoptosis. The results demonstrated that PANX3 was verified as a target gene of miR-367. MiR-367 was found to highly expressed but PANX3, β-catenin, and Wnt5b were observed poorly expressed in fracture mice. downregulated miR-367 increased the mRNA and protein expression of PANX3, β-catenin, and Wnt5b, increased cell growth, proliferation, and migration, while decreased cell apoptosis in osteoblasts. Altogether, our study demonstrates that the downregulation of miR-367 may promote osteoblasts growth and proliferation in fracture through the activation of the PANX3-dependent Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Hong-Lei Jia
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Dong-Sheng Zhou
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
16
|
Fostok SF, El-Sibai M, El-Sabban M, Talhouk RS. Gap Junctions and Wnt Signaling in the Mammary Gland: a Cross-Talk? J Mammary Gland Biol Neoplasia 2019; 24:17-38. [PMID: 30194659 DOI: 10.1007/s10911-018-9411-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/03/2018] [Indexed: 12/21/2022] Open
Abstract
Connexins (Cxs), the building blocks of gap junctions (GJs), exhibit spatiotemporal patterns of expression and regulate the development and differentiation of the mammary gland, acting via channel-dependent and channel-independent mechanisms. Impaired Cx expression and localization are reported in breast cancer, suggesting a tumor suppressive role for Cxs. The signaling events that mediate the role of GJs in the development and tumorigenesis of the mammary gland remain poorly identified. The Wnt pathways, encompassing the canonical or the Wnt/β-catenin pathway and the noncanonical β-catenin-independent pathway, also play important roles in those processes. Indeed, aberrant Wnt signaling is associated with breast cancer. Despite the coincident roles of Cxs and Wnt pathways, the cross-talk in the breast tissue is poorly defined, although this is reported in a number of other tissues. Our previous studies revealed a channel-independent role for Cx43 in inducing differentiation or suppressing tumorigenesis of mammary epithelial cells by acting as a negative regulator of the Wnt/β-catenin pathway. Here, we provide a brief overview of mammary gland development, with emphasis on the role of Cxs in development and tumorigenesis of this tissue. We also discuss the role of Wnt signaling in similar contexts, and review the literature illustrating interplay between Cxs and Wnt pathways.
Collapse
Affiliation(s)
- Sabreen F Fostok
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), P.O. Box: 11-0236, Beirut, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut (AUB), Beirut, Lebanon
| | - Rabih S Talhouk
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), P.O. Box: 11-0236, Beirut, Lebanon.
| |
Collapse
|
17
|
Lopes D, Martins-Cruz C, Oliveira MB, Mano JF. Bone physiology as inspiration for tissue regenerative therapies. Biomaterials 2018; 185:240-275. [PMID: 30261426 PMCID: PMC6445367 DOI: 10.1016/j.biomaterials.2018.09.028] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/15/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022]
Abstract
The development, maintenance of healthy bone and regeneration of injured tissue in the human body comprise a set of intricate and finely coordinated processes. However, an analysis of current bone regeneration strategies shows that only a small fraction of well-reported bone biology aspects has been used as inspiration and transposed into the development of therapeutic products. Specific topics that include inter-scale bone structural organization, developmental aspects of bone morphogenesis, bone repair mechanisms, role of specific cells and heterotypic cell contact in the bone niche (including vascularization networks and immune system cells), cell-cell direct and soluble-mediated contact, extracellular matrix composition (with particular focus on the non-soluble fraction of proteins), as well as mechanical aspects of native bone will be the main reviewed topics. In this Review we suggest a systematic parallelization of (i) fundamental well-established biology of bone, (ii) updated and recent advances on the understanding of biological phenomena occurring in native and injured tissue, and (iii) critical discussion of how those individual aspects have been translated into tissue regeneration strategies using biomaterials and other tissue engineering approaches. We aim at presenting a perspective on unexplored aspects of bone physiology and how they could be translated into innovative regeneration-driven concepts.
Collapse
Affiliation(s)
- Diana Lopes
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago,, 3810 193 Aveiro, Portugal
| | - Cláudia Martins-Cruz
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago,, 3810 193 Aveiro, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago,, 3810 193 Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago,, 3810 193 Aveiro, Portugal.
| |
Collapse
|
18
|
Lee VR, Barr KJ, Kelly JJ, Johnston D, Brown CFC, Robb KP, Sayedyahossein S, Huang K, Gros R, Flynn LE, Penuela S. Pannexin 1 regulates adipose stromal cell differentiation and fat accumulation. Sci Rep 2018; 8:16166. [PMID: 30385873 PMCID: PMC6212408 DOI: 10.1038/s41598-018-34234-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/11/2018] [Indexed: 12/20/2022] Open
Abstract
Pannexin 1 (Panx1) is a channel-forming glycoprotein important in paracrine signaling and cellular development. In this study, we discovered that mice globally lacking Panx1 (KO) have significantly greater total fat mass and reduced lean mass compared to wild type (WT) mice under a normal diet. Despite having higher fat content, Panx1 KO mice on a high fat diet exhibited no differences in weight gain and blood markers of obesity as compared to WT controls, except for an increase in glucose and insulin levels. However, metabolic cage data revealed that these Panx1 KO mice display significantly increased activity levels, higher ambulatory activity, and reduced sleep duration relative to their WT littermates on a high-fat diet. To uncover the cellular mechanism responsible for the increased fat content in the KO, we isolated primary cultures of adipose-derived stromal cells (ASCs) from WT and KO fat pads. In WT ASCs we observed that Panx1 protein levels increase upon induction into an adipogenic lineage. ASCs isolated from Panx1 KO mice proliferate less but demonstrate enhanced adipogenic differentiation with increased intracellular lipid accumulation, glycerol-3-phosphate dehydrogenase (GPDH) enzyme activity, and adipokine secretion, as compared to WT ASCs. This was consistent with the increased adipocyte size and decreased adipocyte numbers observed in subcutaneous fat of the Panx1 KO mice compared to WT. We concluded that Panx1 plays a key role in adipose stromal cells during the early stages of adipogenic proliferation and differentiation, regulating fat accumulation in vivo.
Collapse
Affiliation(s)
- Vanessa R Lee
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A5C1, Canada
| | - Kevin J Barr
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A5C1, Canada
| | - John J Kelly
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A5C1, Canada
| | - Danielle Johnston
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A5C1, Canada
| | - Cody F C Brown
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A5C1, Canada
| | - Kevin P Robb
- Biomedical Engineering Graduate Program, University of Western Ontario, London, Ontario, Canada
| | - Samar Sayedyahossein
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A5C1, Canada
| | - Kenneth Huang
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A5C1, Canada
| | - Robert Gros
- Departments of Physiology and Pharmacology, and of Medicine, University of Western Ontario, London, Ontario, Canada
- Molecular Medicine Research Group Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Lauren E Flynn
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A5C1, Canada
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, Ontario, Canada
| | - Silvia Penuela
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A5C1, Canada.
| |
Collapse
|
19
|
Tanaka T, Hoshijima M, Sunaga J, Nishida T, Hashimoto M, Odagaki N, Osumi R, Aadachi T, Kamioka H. Analysis of Ca 2+ response of osteocyte network by three-dimensional time-lapse imaging in living bone. J Bone Miner Metab 2018; 36:519-528. [PMID: 29027020 DOI: 10.1007/s00774-017-0868-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 08/30/2017] [Indexed: 11/26/2022]
Abstract
Osteocytes form a three-dimensional (3D) cellular network within the mineralized bone matrix. The cellular network has important roles in mechanosensation and mechanotransduction related to bone homeostasis. We visualized the embedded osteocyte network in chick calvariae and observed the flow-induced Ca2+ signaling in osteocytes using 3D time-lapse imaging. In response to the flow, intracellular Ca2+ ([Ca2+]i) significantly increased in developmentally mature osteocytes in comparison with young osteocytes in the bone matrix. To investigate the differences in response between young and developmentally mature osteocytes in detail, we evaluated the expression of osteocyte-related genes using the osteocyte-like cell line MLO-Y4, which was 3D-cultured within type I collagen gels. We found that the c-Fos, Cx43, Panx3, Col1a1, and OCN mRNA levels significantly increased on day 15 in comparison with day 7. These findings indicate that developmentally mature osteocytes are more responsive to mechanical stress than young osteocytes and have important functions in bone formation and remodeling.
Collapse
Affiliation(s)
- Tomoyo Tanaka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 5-1 Shikata-cho, 2-chome, Kitaku, Okayama, 700-8525, Japan
| | - Mitsuhiro Hoshijima
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 5-1 Shikata-cho, 2-chome, Kitaku, Okayama, 700-8525, Japan
- Advanced Research Center for Oral and Craniofacial Sciences, Dental School, Okayama University, Okayama, Japan
| | - Junko Sunaga
- Laboratory of Biomechanics, Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takashi Nishida
- Department of Biochemistry and Molecular Dentistry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mana Hashimoto
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 5-1 Shikata-cho, 2-chome, Kitaku, Okayama, 700-8525, Japan
| | - Naoya Odagaki
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 5-1 Shikata-cho, 2-chome, Kitaku, Okayama, 700-8525, Japan
| | - Ryuta Osumi
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 5-1 Shikata-cho, 2-chome, Kitaku, Okayama, 700-8525, Japan
| | - Taiji Aadachi
- Laboratory of Biomechanics, Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 5-1 Shikata-cho, 2-chome, Kitaku, Okayama, 700-8525, Japan.
| |
Collapse
|
20
|
Connexins and Pannexins: Important Players in Tumorigenesis, Metastasis and Potential Therapeutics. Int J Mol Sci 2018; 19:ijms19061645. [PMID: 29865195 PMCID: PMC6032133 DOI: 10.3390/ijms19061645] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022] Open
Abstract
Since their characterization more than five decades ago, gap junctions and their structural proteins-the connexins-have been associated with cancer cell growth. During that period, the accumulation of data and molecular knowledge about this association revealed an apparent contradictory relationship between them and cancer. It appeared that if gap junctions or connexins can down regulate cancer cell growth they can be also implied in the migration, invasion and metastatic dissemination of cancer cells. Interestingly, in all these situations, connexins seem to be involved through various mechanisms in which they can act either as gap-junctional intercellular communication mediators, modulators of signalling pathways through their interactome, or as hemichannels, which mediate autocrine/paracrine communication. This complex involvement of connexins in cancer progression is even more complicated by the fact that their hemichannel function may overlap with other gap junction-related proteins, the pannexins. Despite this complexity, the possible involvements of connexins and pannexins in cancer progression and the elucidation of the mechanisms they control may lead to use them as new targets to control cancer progression. In this review, the involvements of connexins and pannexins in these different topics (cancer cell growth, invasion/metastasis process, possible cancer therapeutic targets) are discussed.
Collapse
|
21
|
Pham TL, St-Pierre ME, Ravel-Chapuis A, Parks TEC, Langlois S, Penuela S, Jasmin BJ, Cowan KN. Expression of Pannexin 1 and Pannexin 3 during skeletal muscle development, regeneration, and Duchenne muscular dystrophy. J Cell Physiol 2018; 233:7057-7070. [PMID: 29744875 DOI: 10.1002/jcp.26629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 03/30/2018] [Indexed: 01/17/2023]
Abstract
Pannexin 1 (Panx1) and Pannexin 3 (Panx3) are single membrane channels recently implicated in myogenic commitment, as well as myoblast proliferation and differentiation in vitro. However, their expression patterns during skeletal muscle development and regeneration had yet to be investigated. Here, we show that Panx1 levels increase during skeletal muscle development becoming highly expressed together with Panx3 in adult skeletal muscle. In adult mice, Panx1 and Panx3 were differentially expressed in fast- and slow-twitch muscles. We also report that Panx1/PANX1 and Panx3/PANX3 are co-expressed in mouse and human satellite cells, which play crucial roles in skeletal muscle regeneration. Interestingly, Panx1 and Panx3 levels were modulated in muscle degeneration/regeneration, similar to the pattern seen during skeletal muscle development. As Duchenne muscular dystrophy is characterized by skeletal muscle degeneration and impaired regeneration, we next used mild and severe mouse models of this disease and found a significant dysregulation of Panx1 and Panx3 levels in dystrophic skeletal muscles. Together, our results are the first demonstration that Panx1 and Panx3 are differentially expressed amongst skeletal muscle types with their levels being highly modulated during skeletal muscle development, regeneration, and dystrophy. These findings suggest that Panx1 and Panx3 channels may play important and distinct roles in healthy and diseased skeletal muscles.
Collapse
Affiliation(s)
- Tammy L Pham
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Marie-Eve St-Pierre
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Tara E C Parks
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Stéphanie Langlois
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Surgery, Division of Pediatric Surgery, University of Ottawa, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Kyle N Cowan
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Surgery, Division of Pediatric Surgery, University of Ottawa, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
22
|
Morse A, Schindeler A, McDonald MM, Kneissel M, Kramer I, Little DG. Sclerostin Antibody Augments the Anabolic Bone Formation Response in a Mouse Model of Mechanical Tibial Loading. J Bone Miner Res 2018; 33:486-498. [PMID: 29090474 DOI: 10.1002/jbmr.3330] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/11/2017] [Accepted: 10/29/2017] [Indexed: 12/18/2022]
Abstract
Decreased activity or expression of sclerostin, an endogenous inhibitor of Wnt/β-catenin signaling, results in increased bone formation and mass. Antibodies targeting and neutralizing sclerostin (Scl-Ab) have been shown to increase bone mass and reduce fracture risk. Sclerostin is also important in modulating the response of bone to changes in its biomechanical environment. However, the effects of Scl-Ab on mechanotransduction are unclear, and it was speculated that the loading response may be altered for individuals receiving Scl-Ab therapy. To address this, we carried out a 2-week study of tibial cyclic compressive loading on C57Bl/6 mice treated with vehicle or 100 mg/kg/wk Scl-Ab. Increases in bone volume, density, and dynamic bone formation were found with loading, and the anabolic response was further increased by the combination of load and Scl-Ab. To investigate the underlying mechanism, gene profiling by RNA sequencing (RNAseq) was performed on tibias isolated from mice from all four experimental groups. Major alterations in Wnt/β-catenin gene expression were found with tibial loading, however not with Scl-Ab treatment alone. Notably, the combination of load and Scl-Ab elicited a synergistic response from a number of specific Wnt-related and mechanotransduction factors. An unexpected finding was significant upregulation of factors in the Rho GTPase signaling pathway with combination treatment. In summary, combination therapy had a more profound anabolic response than either Scl-Ab or loading treatment alone. The Wnt/β-catenin and Rho GTPase pathways were implicated within bone mechanotransduction and support the concept that bone mechanotransduction is likely to encompass a number of interconnected signaling pathways. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alyson Morse
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Michelle M McDonald
- Bone Biology Program, The Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - David G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
23
|
Hainz N, Beckmann A, Schubert M, Haase A, Martin U, Tschernig T, Meier C. Human stem cells express pannexins. BMC Res Notes 2018; 11:54. [PMID: 29357945 PMCID: PMC5778636 DOI: 10.1186/s13104-018-3125-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/03/2018] [Indexed: 11/23/2022] Open
Abstract
Objective Pannexins are channel proteins important for the release of calcium and adenosine triphosphate, which are among other functions involved in early development. Here, the expression of pannexins was investigated in induced pluripotent stem cells derived from human cord blood endothelial cells (hCBiPS2), in hematopoietic stem cell-derived induced pluripotent stem cells (HSC_F1285_T-iPS2) and in human embryonic stem cells (HES-3). The expression of pannexin (Panx) 1–3 mRNAs was analyzed in all three undifferentiated stem cell lines. Stem cells then underwent undirected differentiation into embryoid bodies and were analyzed regarding expression of germ layer-specific genes. Results Panx1, Panx2, and Panx3 mRNAs were expressed in all undifferentiated stem cell lines investigated. In comparison, Panx1 showed the highest expression among all pannexins. The undirected differentiation resulted in a mixed germ layer genotype in all three stem cell lines. Whereas the expression of Panx1 was not affected by differentiation, the expression of Panx2 was slightly increased in differentiated hCBiPS2 cells, HSC_F1285_T-iPS2 as well as HES3 cells as compared to their undifferentiated counterparts. A slight increase of Panx3 expression was observed in differentiated hCBiPS2 cells only. In conclusion, pluripotent stem cells express all three pannexin genes. Electronic supplementary material The online version of this article (10.1186/s13104-018-3125-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nadine Hainz
- Department of Anatomy and Cell Biology, Saarland University, Kirrberger Straße, Building 61, Saar, 66421, Homburg, Germany
| | - Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, Kirrberger Straße, Building 61, Saar, 66421, Homburg, Germany
| | - Madline Schubert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany
| | - Alexandra Haase
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany
| | - Thomas Tschernig
- Department of Anatomy and Cell Biology, Saarland University, Kirrberger Straße, Building 61, Saar, 66421, Homburg, Germany.
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, Kirrberger Straße, Building 61, Saar, 66421, Homburg, Germany
| |
Collapse
|
24
|
Regulation of Skeletal Muscle Myoblast Differentiation and Proliferation by Pannexins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 925:57-73. [PMID: 27518505 DOI: 10.1007/5584_2016_53] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Pannexins are newly discovered channels that are now recognized as mediators of adenosine triphosphate release from several cell types allowing communication with the extracellular environment. Pannexins have been associated with various physiological and pathological processes including apoptosis, inflammation, and cancer. However, it is only recently that our work has unveiled a role for Pannexin 1 and Pannexin 3 as novel regulators of skeletal muscle myoblast proliferation and differentiation. Myoblast differentiation is an ordered multistep process that includes withdrawal from the cell cycle and the expression of key myogenic factors leading to myoblast differentiation and fusion into multinucleated myotubes. Eventually, myotubes will give rise to the diverse muscle fiber types that build the complex skeletal muscle architecture essential for body movement, postural behavior, and breathing. Skeletal muscle cell proliferation and differentiation are crucial processes required for proper skeletal muscle development during embryogenesis, as well as for the postnatal skeletal muscle regeneration that is necessary for muscle repair after injury or exercise. However, defects in skeletal muscle cell differentiation and/or deregulation of cell proliferation are involved in various skeletal muscle pathologies. In this review, we will discuss the expression of pannexins and their post-translational modifications in skeletal muscle, their known functions in various steps of myogenesis, including myoblast proliferation and differentiation, as well as their possible roles in skeletal muscle development, regeneration, and diseases such as Duchenne muscular dystrophy.
Collapse
|
25
|
Pannexin 3 regulates proliferation and differentiation of odontoblasts via its hemichannel activities. PLoS One 2017; 12:e0177557. [PMID: 28494020 PMCID: PMC5426780 DOI: 10.1371/journal.pone.0177557] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 04/28/2017] [Indexed: 12/25/2022] Open
Abstract
Highly coordinated regulation of cell proliferation and differentiation contributes to the formation of functionally shaped and sized teeth; however, the mechanism underlying the switch from cell cycle exit to cell differentiation during odontogenesis is poorly understood. Recently, we identified pannexin 3 (Panx3) as a member of the pannexin gap junction protein family from tooth germs. The expression of Panx3 was predominately localized in preodontoblasts that arise from dental papilla cells and can differentiate into dentin-secreting odontoblasts. Panx3 also co-localized with p21, a cyclin-dependent kinase inhibitor protein, in preodontoblasts. Panx3 was expressed in primary dental mesenchymal cells and in the mDP dental mesenchymal cell line. Both Panx3 and p21 were induced during the differentiation of mDP cells. Overexpression of Panx3 in mDP cells reduced cell proliferation via up-regulation of p21, but not of p27, and promoted the Bone morphogenetic protein 2 (BMP2)-induced phosphorylation of Smad1/5/8 and the expression of dentin sialophosphoprotein (Dspp), a marker of differentiated odontoblasts. Furthermore, Panx3 released intracellular ATP into the extracellular space through its hemichannel and induced the phosphorylation of AMP-activated protein kinase (AMPK). 5-Aminoimidazole-4-carboxamide-ribonucleoside (AICAR), an activator of AMPK, reduced mDP cell proliferation and induced p21 expression. Conversely, knockdown of endogenous Panx3 by siRNA inhibited AMPK phosphorylation, p21 expression, and the phosphorylation of Smad1/5/8 even in the presence of BMP2. Taken together, our results suggest that Panx3 modulates intracellular ATP levels, resulting in the inhibition of odontoblast proliferation through the AMPK/p21 signaling pathway and promotion of cell differentiation by the BMP/Smad signaling pathway.
Collapse
|
26
|
Dorotheou D, Farsadaki V, Bochaton-Piallat ML, Giannopoulou C, Halazonetis TD, Kiliaridis S. Increased Cell Proliferation and Gene Expression of Genes Related to Bone Remodeling, Cell Adhesion and Collagen Metabolism in the Periodontal Ligament of Unopposed Molars in Growing Rats. Front Physiol 2017; 8:75. [PMID: 28239357 PMCID: PMC5301028 DOI: 10.3389/fphys.2017.00075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/26/2017] [Indexed: 01/10/2023] Open
Abstract
Tooth eruption, the process by which teeth emerge from within the alveolar bone into the oral cavity, is poorly understood. The post-emergent phase of tooth eruption continues throughout life, in particular, if teeth are not opposed by antagonists. The aim of the present study was to better understand the molecular processes underlying post-emergent tooth eruption. Toward this goal, we removed the crowns of the maxillary molars on one side of the mouth of 14 young rats and examined gene expression patterns in the periodontal ligaments (PDLs) of the ipsilateral and contralateral mandibular molars, 3 and 15 days later. Nine untreated rats served as controls. Expression of six genes, Adamts18, Ostn, P4ha3, Panx3, Pth1r, and Tnmd, was upregulated in unopposed molars relative to molars with antagonists. These genes function in osteoblast differentiation and proliferation, cell adhesion and collagen metabolism. Proliferation of PDL cells also increased following loss of the antagonist teeth. Interestingly, mutations in PTH1R have been linked to defects in the post-emergent phase of tooth eruption in humans. We conclude that post-emergent eruption of unopposed teeth is associated with gene expression patterns conducive to alveolar bone formation and PDL remodeling.
Collapse
Affiliation(s)
- Domna Dorotheou
- Department of Orthodontics, University of Geneva Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Fifty years ago, tumour cells were found to lack electrical coupling, leading to the hypothesis that loss of direct intercellular communication is commonly associated with cancer onset and progression. Subsequent studies linked this phenomenon to gap junctions composed of connexin proteins. Although many studies support the notion that connexins are tumour suppressors, recent evidence suggests that, in some tumour types, they may facilitate specific stages of tumour progression through both junctional and non-junctional signalling pathways. This Timeline article highlights the milestones connecting gap junctions to cancer, and underscores important unanswered questions, controversies and therapeutic opportunities in the field.
Collapse
Affiliation(s)
- Trond Aasen
- (Co-corresponding authors) Correspondence to
T.A. () and D.W.L.
()
| | - Marc Mesnil
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences
Fondamentales et Appliquées, Université de Poitiers, Poitiers,
France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, The Life
Sciences Institute, University of British Columbia, Vancouver, British
Columbia, Canada
| | - Paul D. Lampe
- Translational Research Program, Fred Hutchinson Cancer Research
Center, Seattle, United States
| | - Dale W. Laird
- (Co-corresponding authors) Correspondence to
T.A. () and D.W.L.
()
| |
Collapse
|
28
|
Abstract
Cell-cell and cell-matrix communications play important roles in both cell proliferation and differentiation. Gap junction proteins mediate signaling communication by exchanging small molecules and dramatically stimulating intracellular signaling pathways to determine cell fate. Vertebrates have 2 gap junction families: pannexins (Panxs) and connexins (Cxs). Unlike Cxs, the functions of Panxs are not fully understood. In skeletal formation, Panx3 and Cx43 are the most abundantly expressed gap junction proteins from each family. Panx3 is induced in the transient stage from the proliferation and differentiation of chondrocytes and osteoprogenitor cells. Panx3 regulates both chondrocyte and osteoblast differentiation via the activation of intracellular Ca2+ signaling pathways through multiple channel activities: hemichannels, endoplasmic reticulum (ER) Ca2+ channels, and gap junctions. Moreover, Panx3 also inhibits osteoprogenitor cell proliferation and promotes cell cycle exit through the inactivation of Wnt/β-catenin signaling and the activation of p21. Panx3-knockout (KO) mice have more severe skeletal abnormalities than those of Cx43-KO mice. A phenotypic analysis of Panx3-KO mice indicates that Panx3 regulates the terminal differentiation of chondrocytes by promoting vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP) 13. Based on the generation of Panx3-/-; Cx43-/- mice, Panx3 is upstream of Cx43 in osteogenesis. Panx3 promotes Cx43 expression by regulating Wnt/β-catenin signaling and osterix expression. Further, although Panx3 can function in 3 ways, Cx43 cannot function through the ER Ca2+ channel, only via the hemichannels and gap junction routes. In this review, we discuss the current knowledge regarding the roles of Panx3 in skeletal formation and address the potential for new therapies in the treatment of diseases and pathologies associated with Panx3, such as osteoarthritis (OA).
Collapse
Affiliation(s)
- M Ishikawa
- 1 Division of Operative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan.,2 Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Y Yamada
- 2 Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
29
|
Differential effects of pannexins on noise-induced hearing loss. Biochem J 2016; 473:4665-4680. [PMID: 27784763 DOI: 10.1042/bcj20160668] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/14/2016] [Accepted: 10/24/2016] [Indexed: 12/20/2022]
Abstract
Hearing loss, including noise-induced hearing loss, is highly prevalent and severely hinders an individual's quality of life, yet many of the mechanisms that cause hearing loss are unknown. The pannexin (Panx) channel proteins, Panx1 and Panx3, are regionally expressed in many cell types along the auditory pathway, and mice lacking Panx1 in specific cells of the inner ear exhibit hearing loss, suggesting a vital role for Panxs in hearing. We proposed that Panx1 and/or Panx3 null mice would exhibit severe hearing loss and increased susceptibility to noise-induced hearing loss. Using the auditory brainstem response, we surprisingly found that Panx1-/- and Panx3-/- mice did not harbor hearing or cochlear nerve deficits. Furthermore, while Panx1-/- mice displayed no protection against loud noise-induced hearing loss, Panx3-/- mice exhibited enhanced 16- and 24-kHz hearing recovery 7 days after a loud noise exposure (NE; 12 kHz tone, 115 dB sound pressure level, 1 h). Interestingly, Cx26, Cx30, Cx43, and Panx2 were up-regulated in Panx3-/- mice compared with wild-type and/or Panx1-/- mice, and assessment of the auditory tract revealed morphological changes in the middle ear bones of Panx3-/- mice. It is unclear if these changes alone are sufficient to provide protection against loud noise-induced hearing loss. Contrary to what we expected, these data suggest that Panx1 and Panx3 are not essential for baseline hearing in mice tested, but the therapeutic targeting of Panx3 may prove protective against mid-high-frequency hearing loss caused by loud NE.
Collapse
|
30
|
Song F, Sun H, Wang Y, Yang H, Huang L, Fu D, Gan J, Huang C. Pannexin3 inhibits TNF-α-induced inflammatory response by suppressing NF-κB signalling pathway in human dental pulp cells. J Cell Mol Med 2016; 21:444-455. [PMID: 27679980 PMCID: PMC5323855 DOI: 10.1111/jcmm.12988] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/16/2016] [Indexed: 01/21/2023] Open
Abstract
Human dental pulp cells (HDPCs) play a crucial role in dental pulp inflammation. Pannexin 3 (Panx3), a member of Panxs (Pannexins), has been recently found to be involved in inflammation. However, the mechanism of Panx3 in human dental pulp inflammation remains unclear. In this study, the role of Panx3 in inflammatory response was firstly explored, and its potential mechanism was proposed. Immunohistochemical staining showed that Panx3 levels were diminished in inflamed human and rat dental pulp tissues. In vitro, Panx3 expression was significantly down‐regulated in HDPCs following a TNF‐α challenge in a concentration‐dependent way, which reached the lowest level at 10 ng/ml of TNF‐α. Such decrease could be reversed by MG132, a proteasome inhibitor. Unlike MG132, BAY 11‐7082, a NF‐κB inhibitor, even reinforced the inhibitory effect of TNF‐α. Quantitative real‐time PCR (qRT‐PCR) and enzyme‐linked immunosorbent assay (ELISA) were used to investigate the role of Panx3 in inflammatory response of HDPCs. TNF‐α‐induced pro‐inflammatory cytokines, interleukin (IL)‐1β and IL‐6, were significantly lessened when Panx3 was overexpressed in HDPCs. Conversely, Panx3 knockdown exacerbated the expression of pro‐inflammatory cytokines. Moreover, Western blot, dual‐luciferase reporter assay, immunofluorescence staining, qRT‐PCR and ELISA results showed that Panx3 participated in dental pulp inflammation in a NF‐κB‐dependent manner. These findings suggested that Panx3 has a defensive role in dental pulp inflammation, serving as a potential target to be exploited for the intervention of human dental pulp inflammation.
Collapse
Affiliation(s)
- Fangfang Song
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Hualing Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Yake Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Hongye Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Liyuan Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Dongjie Fu
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Gan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Cui Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
31
|
Halliwill KD, Quigley DA, Kang HC, Del Rosario R, Ginzinger D, Balmain A. Panx3 links body mass index and tumorigenesis in a genetically heterogeneous mouse model of carcinogen-induced cancer. Genome Med 2016; 8:83. [PMID: 27506198 PMCID: PMC4977876 DOI: 10.1186/s13073-016-0334-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/11/2016] [Indexed: 01/01/2023] Open
Abstract
Background Body mass index (BMI) has been implicated as a primary factor influencing cancer development. However, understanding the relationship between these two complex traits has been confounded by both environmental and genetic heterogeneity. Methods In order to gain insight into the genetic factors linking BMI and cancer, we performed chemical carcinogenesis on a genetically heterogeneous cohort of interspecific backcross mice ((Mus Spretus × FVB/N) F1 × FVB/N). Using this cohort, we performed quantitative trait loci (QTL) analysis to identify regions linked to BMI. We then performed an integrated analysis incorporating gene expression, sequence comparison between strains, and gene expression network analysis to identify candidate genes influencing both tumor development and BMI. Results Analysis of QTL linked to tumorigenesis and BMI identified several loci associated with both phenotypes. Exploring these loci in greater detail revealed a novel relationship between the Pannexin 3 gene (Panx3) and both BMI and tumorigenesis. Panx3 is positively associated with BMI and is strongly tied to a lipid metabolism gene expression network. Pre-treatment Panx3 gene expression levels in normal skin are associated with tumor susceptibility and inhibition of Panx function strongly influences inflammation. Conclusions These studies have identified several genetic loci that influence both BMI and carcinogenesis and implicate Panx3 as a candidate gene that links these phenotypes through its effects on inflammation and lipid metabolism. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0334-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kyle D Halliwill
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - David A Quigley
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Hio Chung Kang
- Invitae Corporation, 458 Brannan St, San Francisco, CA, 94107, USA
| | - Reyno Del Rosario
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - David Ginzinger
- Thermo Fisher Scientific, 5791 Van Allen Way, Carlsbad, CA, 92008, USA
| | - Allan Balmain
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA. .,Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
| |
Collapse
|
32
|
Bond SR, Abramyan J, Fu K, Naus CC, Richman JM. Pannexin 3 is required for late stage bone growth but not for initiation of ossification in avian embryos. Dev Dyn 2016; 245:913-24. [PMID: 27295565 DOI: 10.1002/dvdy.24425] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 05/23/2016] [Accepted: 05/29/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Pannexin 3 (PANX3) is a channel-forming protein capable of stimulating osteogenesis in vitro. Here, we studied the in vivo roles of PANX3 in the chicken embryo using the RCAS retroviral system to over-express and knockdown expression during endochondral bone formation. RESULTS In the limbs, PANX3 RNA was first detected in the cartilage condensations and became restricted to the prehypertrophic cartilage of the epiphyses, diaphysis, and perichondrium. The increase in PANX3 was not sufficient to alter osteogenesis; however, knockdown with a virus containing an interference RNA construct caused a 20% reduction in bone volume. The control virus containing an shEGFP cassette did not affect development. Interestingly, the phenotype was restricted to later stages rather than to proliferation of the skeletogenic mesenchyme, formation of the cartilage condensation, or creation of the hypertrophic zones. In addition, there was also no change in readouts of Hedgehog, WNT, fibroblast growth factor, or bone morphogenetic protein signaling using either quantitative real-time polymerase chain reaction or radioactive in situ hybridization. CONCLUSIONS Based on the normal expression domains of PANX3 and the relatively late manifestation of the phenotype, it is possible that PANX3 hemichannels may be required to facilitate the transition of hypertrophic chondrocytes to osteoblasts, thereby achieving final bone size. Developmental Dynamics 245:913-924, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Stephen R Bond
- Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - John Abramyan
- Faculty of Dentistry, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kathy Fu
- Faculty of Dentistry, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christian C Naus
- Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joy M Richman
- Faculty of Dentistry, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
33
|
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: 3.8] [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
|
34
|
Ishikawa M, Williams GL, Ikeuchi T, Sakai K, Fukumoto S, Yamada Y. Pannexin 3 and connexin 43 modulate skeletal development through their distinct functions and expression patterns. J Cell Sci 2016; 129:1018-30. [PMID: 26759176 DOI: 10.1242/jcs.176883] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/05/2016] [Indexed: 01/24/2023] Open
Abstract
Pannexin 3 (Panx3) and connexin 43 (Cx43; also known as GJA1) are two major gap junction proteins expressed in osteoblasts. Here, we studied their functional relationships in skeletal formation by generating Panx3(-/-) and Panx3(-/-);Cx43(-/-) mice and comparing their skeletal phenotypes with Cx43(-/-) mice. Panx3(-/-) mice displayed defects in endochondral and intramembranous ossification, resulting in severe dwarfism and reduced bone density. The skeletal abnormalities of Panx3(-/-);Cx43(-/-) mice were similar to those in Panx3(-/-) mice. The gross appearance of newborn Cx43(-/-) skeletons showed no obvious abnormalities, except for less mineralization of the skull. In Panx3(-/-) mice, proliferation of chondrocytes and osteoblasts increased and differentiation of these cells was inhibited. Panx3 promoted expression of osteogenic proteins such as ALP and Ocn (also known as ALPL and BGLAP, respectively), as well as Cx43, by regulating Osx (also known as SP7) expression. Panx3 was induced in the early differentiation stage and reduced during the maturation stage of osteoblasts, when Cx43 expression increased in order to promote mineralization. Furthermore, only Panx3 functioned as an endoplasmic reticulum (ER) Ca(2+) channel to promote differentiation, and it could rescue mineralization defects in Cx43(-/-) calvarial cells. Our findings reveal that Panx3 and Cx43 have distinct functions in skeletal formation.
Collapse
Affiliation(s)
- Masaki Ishikawa
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20814, USA Operative Dentistry, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Geneva L Williams
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20814, USA Division of Biology & Biomedical Sciences, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Tomoko Ikeuchi
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20814, USA
| | - Kiyoshi Sakai
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20814, USA Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Satoshi Fukumoto
- Department of Pediatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai 980-8576, Japan
| | - Yoshihiko Yamada
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20814, USA
| |
Collapse
|
35
|
Caskenette D, Penuela S, Lee V, Barr K, Beier F, Laird DW, Willmore KE. Global deletion of Panx3 produces multiple phenotypic effects in mouse humeri and femora. J Anat 2016; 228:746-56. [PMID: 26749194 DOI: 10.1111/joa.12437] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2015] [Indexed: 02/04/2023] Open
Abstract
Pannexins form single-membrane channels that allow passage of small molecules between the intracellular and extracellular compartments. Of the three pannexin family members, Pannexin3 (Panx3) is the least studied but is highly expressed in skeletal tissues and is thought to play a role in the regulation of chondrocyte and osteoblast proliferation and differentiation. The purpose of our study is to closely examine the in vivo effects of Panx3 ablation on long bone morphology using micro-computed tomography. Using Panx3 knockout (KO) and wildtype (WT) adult mice, we measured and compared aspects of phenotypic shape, bone mineral density (BMD), cross-sectional geometric properties of right femora and humeri, and lean mass. We found that KO mice have absolutely and relatively shorter diaphyseal shafts compared with WT mice, and relatively larger areas of muscle attachment sites. No differences in BMD or lean mass were found between WT and KO mice. Interestingly, KO mice had more robust femora and humeri compared with WT mice when assessed in cross-section at the midshaft. Our results clearly show that Panx3 ablation produces phenotypic effects in mouse femora and humeri, and support the premise that Panx3 has a role in regulating long bone growth and development.
Collapse
Affiliation(s)
- Deidre Caskenette
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Vanessa Lee
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Kevin Barr
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, London, ON, Canada
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Katherine E Willmore
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
36
|
Yamada A, Futagi M, Fukumoto E, Saito K, Yoshizaki K, Ishikawa M, Arakaki M, Hino R, Sugawara Y, Ishikawa M, Naruse M, Miyazaki K, Nakamura T, Fukumoto S. Connexin 43 Is Necessary for Salivary Gland Branching Morphogenesis and FGF10-induced ERK1/2 Phosphorylation. J Biol Chem 2015; 291:904-12. [PMID: 26565022 DOI: 10.1074/jbc.m115.674663] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 11/06/2022] Open
Abstract
Cell-cell interaction via the gap junction regulates cell growth and differentiation, leading to formation of organs of appropriate size and quality. To determine the role of connexin43 in salivary gland development, we analyzed its expression in developing submandibular glands (SMGs). Connexin43 (Cx43) was found to be expressed in salivary gland epithelium. In ex vivo organ cultures of SMGs, addition of the gap junctional inhibitors 18α-glycyrrhetinic acid (18α-GA) and oleamide inhibited SMG branching morphogenesis, suggesting that gap junctional communication contributes to salivary gland development. In Cx43(-/-) salivary glands, submandibular and sublingual gland size was reduced as compared with those from heterozygotes. The expression of Pdgfa, Pdgfb, Fgf7, and Fgf10, which induced branching of SMGs in Cx43(-/-) samples, were not changed as compared with those from heterozygotes. Furthermore, the blocking peptide for the hemichannel and gap junction channel showed inhibition of terminal bud branching. FGF10 induced branching morphogenesis, while it did not rescue the Cx43(-/-) phenotype, thus Cx43 may regulate FGF10 signaling during salivary gland development. FGF10 is expressed in salivary gland mesenchyme and regulates epithelial proliferation, and was shown to induce ERK1/2 phosphorylation in salivary epithelial cells, while ERK1/2 phosphorylation in HSY cells was dramatically inhibited by 18α-GA, a Cx43 peptide or siRNA. On the other hand, PDGF-AA and PDGF-BB separately induced ERK1/2 phosphorylation in primary cultured salivary mesenchymal cells regardless of the presence of 18α-GA. Together, our results suggest that Cx43 regulates FGF10-induced ERK1/2 phosphorylation in salivary epithelium but not in mesenchyme during the process of SMG branching morphogenesis.
Collapse
Affiliation(s)
- Aya Yamada
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Masaharu Futagi
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Emiko Fukumoto
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Kan Saito
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Keigo Yoshizaki
- Division of Orthodontics, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Masaki Ishikawa
- Operative Dentistry, Department of Restorative Dentistry Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan and
| | - Makiko Arakaki
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Ryoko Hino
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Yu Sugawara
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Momoko Ishikawa
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Masahiro Naruse
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Kanako Miyazaki
- Division of Orthodontics, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Nakamura
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Satoshi Fukumoto
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences,
| |
Collapse
|
37
|
Choi H, Kim TH, Yun CY, Kim JW, Cho ES. Testicular acid phosphatase induces odontoblast differentiation and mineralization. Cell Tissue Res 2015; 364:95-103. [DOI: 10.1007/s00441-015-2310-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/30/2015] [Indexed: 01/01/2023]
|
38
|
Plotkin LI, Stains JP. Connexins and pannexins in the skeleton: gap junctions, hemichannels and more. Cell Mol Life Sci 2015; 72:2853-67. [PMID: 26091748 PMCID: PMC4503509 DOI: 10.1007/s00018-015-1963-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 06/11/2015] [Indexed: 10/23/2022]
Abstract
Regulation of bone homeostasis depends on the concerted actions of bone-forming osteoblasts and bone-resorbing osteoclasts, controlled by osteocytes, cells derived from osteoblasts surrounded by bone matrix. The control of differentiation, viability and function of bone cells relies on the presence of connexins. Connexin43 regulates the expression of genes required for osteoblast and osteoclast differentiation directly or by changing the levels of osteocytic genes, and connexin45 may oppose connexin43 actions in osteoblastic cells. Connexin37 is required for osteoclast differentiation and its deletion results in increased bone mass. Less is known on the role of connexins in cartilage, ligaments and tendons. Connexin43, connexin45, connexin32, connexin46 and connexin29 are expressed in chondrocytes, while connexin43 and connexin32 are expressed in ligaments and tendons. Similarly, although the expression of pannexin1, pannexin2 and pannexin3 has been demonstrated in bone and cartilage cells, their function in these tissues is not fully understood.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., MS 5035, Indianapolis, IN, 46202, USA,
| | | |
Collapse
|
39
|
Oh SK, Shin JO, Baek JI, Lee J, Bae JW, Ankamerddy H, Kim MJ, Huh TL, Ryoo ZY, Kim UK, Bok J, Lee KY. Pannexin 3 is required for normal progression of skeletal development in vertebrates. FASEB J 2015; 29:4473-84. [PMID: 26183770 DOI: 10.1096/fj.15-273722] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/30/2015] [Indexed: 12/25/2022]
Abstract
The vertebrate skeletal system has various functions, including support, movement, protection, and the production of blood cells. The development of cartilage and bones, the core components of the skeletal system, is mediated by systematic inter- and intracellular communication among multiple signaling pathways in differentiating progenitors and the surrounding tissues. Recently, Pannexin (Panx) 3 has been shown to play important roles in bone development in vitro by mediating multiple signaling pathways, although its roles in vivo have not been explored. In this study, we generated and analyzed Panx3 knockout mice and examined the skeletal phenotypes of panx3 morphant zebrafish. Panx3(-/-) embryos exhibited delays in hypertrophic chondrocyte differentiation and osteoblast differentiation as well as the initiation of mineralization, resulting in shortened long bones in adulthood. The abnormal progression of hypertrophic chondrogenesis appeared to be associated with the sustained proliferation of chondrocytes, which resulted from increased intracellular cAMP levels. Similarly, osteoblast differentiation and mineralization were delayed in panx3 morphant zebrafish. Taken together, our results provide evidence of the crucial roles of Panx3 in vertebrate skeletal development in vivo.
Collapse
Affiliation(s)
- Se-Kyung Oh
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeong-Oh Shin
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeong-In Baek
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jinwook Lee
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jae Woong Bae
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harinarayana Ankamerddy
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Myoung-Jin Kim
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tae-Lin Huh
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zae-Young Ryoo
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Un-Kyung Kim
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jinwoong Bok
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kyu-Yup Lee
- *Department of Biology, College of Natural Sciences, School of Life Sciences, BrainKorea21 PLUS Project for Kyungpook National University Creative BioResearch Group, School of Life Science and Biotechnology, and Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Kyungpook National University, Daegu, South Korea; Department of Anatomy, BrainKorea21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea; and Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
40
|
Aasen T. Connexins: junctional and non-junctional modulators of proliferation. Cell Tissue Res 2014; 360:685-99. [PMID: 25547217 DOI: 10.1007/s00441-014-2078-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 11/14/2014] [Indexed: 12/11/2022]
Abstract
Mounting evidence indicates that dysregulation of gap junctions and their structural subunits-connexins-often occurs in, and sometimes causes, a variety of proliferative disorders, including cancer. Connexin-mediated regulation of cell proliferation is complex and may involve modulation of gap junction intercellular communication (GJIC), hemichannel signalling, or gap junction-independent paths. However, the exact mechanisms linking connexins to proliferation remain poorly defined and a number of contradictory studies report both pro- and anti-proliferative effects, effects that often depend on the cell or tissue type or the microenvironment. The present review covers junctional and non-junctional regulation of proliferation by connexins, with a particular emphasis on their association with cancer.
Collapse
Affiliation(s)
- Trond Aasen
- Molecular Pathology Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, Barcelona, 08035, Spain,
| |
Collapse
|
41
|
Langlois S, Xiang X, Young K, Cowan BJ, Penuela S, Cowan KN. Pannexin 1 and pannexin 3 channels regulate skeletal muscle myoblast proliferation and differentiation. J Biol Chem 2014; 289:30717-30731. [PMID: 25239622 PMCID: PMC4215249 DOI: 10.1074/jbc.m114.572131] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pannexins constitute a family of three glycoproteins (Panx1, -2, and -3) forming single membrane channels. Recent work demonstrated that Panx1 is expressed in skeletal muscle and involved in the potentiation of contraction. However, Panxs functions in skeletal muscle cell differentiation, and proliferation had yet to be assessed. We show here that Panx1 and Panx3, but not Panx2, are present in human and rodent skeletal muscle, and their various species are differentially expressed in fetal versus adult human skeletal muscle tissue. Panx1 levels were very low in undifferentiated human primary skeletal muscle cells and myoblasts (HSMM) but increased drastically during differentiation and became the main Panx expressed in differentiated cells. Using HSMM, we found that Panx1 expression promotes this process, whereas it was impaired in the presence of probenecid or carbenoxolone. As for Panx3, its lower molecular weight species were prominent in adult skeletal muscle but very low in the fetal tissue and in undifferentiated skeletal muscle cells and myoblasts. Its overexpression (∼43-kDa species) induced HSMM differentiation and also inhibited their proliferation. On the other hand, a ∼70-kDa immunoreactive species of Panx3, likely glycosylated, sialylated, and phosphorylated, was highly expressed in proliferative myoblasts but strikingly down-regulated during their differentiation. Reduction of its endogenous expression using two Panx3 shRNAs significantly inhibited HSMM proliferation without triggering their differentiation. In summary, our results demonstrate that Panx1 and Panx3 are co-expressed in human skeletal muscle myoblasts and play a pivotal role in dictating the proliferation and differentiation status of these cells.
Collapse
Affiliation(s)
- Stéphanie Langlois
- Department of Surgery, Division of Paediatric Surgery, University of Ottawa, Children's Hospital of Eastern Ontario, Ottawa, Ontario K1H 8L1, Canada,; Apoptosis Research Center, Children's Hospital of Eastern Ontario, Ottawa, Ontario K1H 8L1, Canada
| | - Xiao Xiang
- Apoptosis Research Center, Children's Hospital of Eastern Ontario, Ottawa, Ontario K1H 8L1, Canada,; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada
| | - Kelsey Young
- Apoptosis Research Center, Children's Hospital of Eastern Ontario, Ottawa, Ontario K1H 8L1, Canada
| | - Bryce J Cowan
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada, and
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Kyle N Cowan
- Department of Surgery, Division of Paediatric Surgery, University of Ottawa, Children's Hospital of Eastern Ontario, Ottawa, Ontario K1H 8L1, Canada,; Apoptosis Research Center, Children's Hospital of Eastern Ontario, Ottawa, Ontario K1H 8L1, Canada,; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada,.
| |
Collapse
|
42
|
CREB is activated in smooth muscle cells isolated from atherosclerotic plaques and reduces smooth muscle cell proliferation via p21-dependent mechanism. Int J Cardiol 2014; 174:764-7. [DOI: 10.1016/j.ijcard.2014.04.078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 04/04/2014] [Indexed: 11/22/2022]
|
43
|
Taylor KA, Wright JR, Vial C, Evans RJ, Mahaut-Smith MP. Amplification of human platelet activation by surface pannexin-1 channels. J Thromb Haemost 2014; 12:987-98. [PMID: 24655807 PMCID: PMC4238786 DOI: 10.1111/jth.12566] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 03/04/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND Pannexin-1 (Panx1) forms an anion-selective channel with a permeability up to ~1 kDa and represents a non-lytic, non-vesicular ATP release pathway in erythrocytes, leukocytes and neurons. Related connexin gap junction proteins have been reported in platelets; however, the expression and function of the pannexins remain unknown. OBJECTIVE To determine the expression and function of pannexins in human plate-lets, using molecular, cellular and functional techniques. METHODS Panx1 expression in human platelets was det-ermined using qPCR and antibody-based techniques. Contributions of Panx1 to agonist-evoked efflux of cytoplasmic calcein, Ca(2+) influx, ATP release and aggregation were assessed in washed platelets under conditions where the P2X1 receptor response was preserved (0.32 U mL(-1) apyrase). Thrombus formation in whole blood was assessed in vitro using a shear chamber assay. Two structurally unrelated and widely used Panx1 inhibitors, probenecid and carbenoxolone, were used throughout this study, at concentrations that do not affect connexin channels. RESULTS PANX1, but not PANX2 or PANX3, mRNA was detected in human platelets. Furthermore, Panx1 protein is glycosylated and present on the plasma membrane of platelets, and displays weak physical association with P2X1 receptors. Panx1 inhibition blocked thrombin-evoked efflux of calcein, and reduced Ca(2+) influx, ATP release, platelet aggregation and thrombus formation under arterial shear rates in vitro. The Panx1-dependent contribution was not additive to that of P2X1 receptors. CONCLUSIONS Panx1 is expressed on human platelets and amplifies Ca(2+) influx, ATP release and aggregation through the secondary activation of P2X1 receptors. We propose that Panx1 represents a novel target for the management of arterial thrombosis.
Collapse
Affiliation(s)
- K A Taylor
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | | | | | | | | |
Collapse
|