1
|
Kohama T, Tomioka I, Morohaku K. In vitro production of viable eggs from undeveloped oocytes in mouse preantral follicles by reconstructing granulosa cell-oocyte complexes†. Biol Reprod 2024:ioae125. [PMID: 39237319 DOI: 10.1093/biolre/ioae125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
In vitro culture of ungrown oocytes in preantral follicles is one of the intriguing subjects being pursued to produce viable eggs in assisted reproductive technology. Previous studies have succeeded in obtaining mature eggs after in vitro culture of preantral follicles, while denuded undeveloped oocytes, which are obtained occasionally when collecting preantral follicles, seem to be almost useless. Moreover, methods to culture them efficiently to produce viable eggs have not been established yet. The present study was conducted to demonstrate in vitro culture of mouse denuded undeveloped oocytes by reconstructing granulosa cell-oocyte complexes, and to analyze cellular communication in reconstructed granulosa cell-oocyte complexes. Single denuded undeveloped oocytes were aggregated with 1 × 104 granulosa cells in wells with U-shaped bottoms in a low-binding cell culture plate for 8 days under either 20% or 5% O2, and then the reconstructed granulosa cell-oocyte complexes formed were cultured on a collagen-coated culture membrane insert for 4 days under 5% O2. At day 8 of culture, the rates of reconstructed granulosa cell-oocyte complexes formation were significantly higher in the culture group under 5% O2 (64.9%) than that under 20% O2 (42.3%; P < 0.001); furthermore, the formation of transzonal projections was observed. After maturation and fertilization, we produced matured eggs and blastocysts at higher rates (>90% and 61.9%, respectively) in the group cultured under 5% O2. After transferring 126 two- to four-cell stage embryos, six live pups were obtained. This is the first report that demonstrates production of viable eggs after in vitro culture of denuded undeveloped oocytes from preantral follicles by reconstruction of granulosa cell-oocyte complexes.
Collapse
Affiliation(s)
- Tomohiro Kohama
- Laboratory of Germ Cell Physiology and Engineering, Faculty of Agriculture, Shinshu University, Nagano, Japan
| | - Ikuo Tomioka
- Laboratory of Applied Reproductive Science, Faculty of Agriculture, Shinshu University, Nagano, Japan
| | - Kanako Morohaku
- Laboratory of Germ Cell Physiology and Engineering, Faculty of Agriculture, Shinshu University, Nagano, Japan
| |
Collapse
|
2
|
Cao H, Li L, Liu S, Wang Y, Liu X, Yang F, Dong W. The multifaceted role of extracellular ATP in sperm function: From spermatogenesis to fertilization. Theriogenology 2024; 214:98-106. [PMID: 37865020 DOI: 10.1016/j.theriogenology.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/09/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023]
Abstract
Extracellular adenosine 5'-triphosphate (ATP) is a vital signaling molecule involved in various physiological processes within the body. In recent years, studies have revealed its significant role in male reproduction, particularly in sperm function. This review explores the multifaceted role of extracellular ATP in sperm function, from spermatogenesis to fertilization. We discuss the impact of extracellular ATP on spermatogenesis, sperm maturation and sperm-egg fusion, highlighting the complex regulatory mechanisms and potential clinical applications in the context of male infertility. By examining the latest research, we emphasize the crucial role of extracellular ATP in sperm function and propose future research directions to further.
Collapse
Affiliation(s)
- Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Long Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xianglin Liu
- College of Forestry, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Fangxia Yang
- College of Forestry, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
3
|
Mezache L, Soltisz AM, Johnstone SR, Isakson BE, Veeraraghavan R. Vascular Endothelial Barrier Protection Prevents Atrial Fibrillation by Preserving Cardiac Nanostructure. JACC Clin Electrophysiol 2023; 9:2444-2458. [PMID: 38032579 PMCID: PMC11134328 DOI: 10.1016/j.jacep.2023.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/20/2023] [Accepted: 10/21/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Atrial fibrillation (AF), the most common cardiac arrhythmia, is widely associated with inflammation, vascular dysfunction, and elevated levels of the vascular leak-inducing cytokine, vascular endothelial growth factor (VEGF). Mechanisms underlying AF are poorly understood and current treatments only manage this progressive disease, rather than arresting the underlying pathology. The authors previously identified edema-induced disruption of sodium channel (NaV1.5)-rich intercalated disk nanodomains as a novel mechanism for AF initiation secondary to acute inflammation. Therefore, we hypothesized that protecting the vascular barrier can prevent vascular leak-induced atrial arrhythmias. OBJECTIVES In this study the authors tested the hypothesis that protecting the vascular barrier can prevent vascular leak-induced atrial arrhythmias. They identified 2 molecular targets for vascular barrier protection, connexin43 (Cx43) hemichannels and pannexin-1 (Panx1) channels, which have been implicated in cytokine-induced vascular leak. METHODS The authors undertook in vivo electrocardiography, electron microscopy, and super-resolution light microscopy studies in mice acutely treated with a clinically relevant level of VEGF. RESULTS AF incidence was increased in untreated mice exposed to VEGF relative to vehicle control subjects. VEGF also increased the average number of AF episodes. VEGF shifted NaV1.5 signal to longer distances from Cx43 gap junctions, measured by a distance transformation-based spatial analysis of 3-dimensional confocal images of intercalated disks. Similar effects were observed with NaV1.5 localized near mechanical junctions composed of neural cadherin. Blocking connexin43 hemichannels (αCT11 peptide) or Panx1 channels (PxIL2P peptide) significantly reduced the duration of AF episodes compared with VEGF alone with no treatment. Concurrently, both peptide therapies preserved NaV1.5 distance from gap junctions to control levels and reduced mechanical junction-adjacent intermembrane distance in these hearts. Notably, similar antiarrhythmic efficacy was also achieved with clinically-relevant small-molecule inhibitors of Cx43 and Panx1. CONCLUSIONS These results highlight vascular barrier protection as an antiarrhythmic strategy following inflammation-induced vascular leak.
Collapse
Affiliation(s)
- Louisa Mezache
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Andrew M Soltisz
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Scott R Johnstone
- Fralin Biomedical Research Institute at VTC, Centre for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA; Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA; Virginia Tech Carilion School of Medicine, Department of Surgery, Roanoke, Virginia, USA
| | - Brant E Isakson
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, Virginia, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
4
|
Lucaciu SA, Figliuzzi R, Neumann R, Nazarali S, Del Sordo L, Leighton SE, Hauser A, Shao Q, Johnston D, Bai D, Laird DW. GJB4 variants linked to skin disease exhibit a trafficking deficiency en route to gap junction formation that can be restored by co-expression of select connexins. Front Cell Dev Biol 2023; 11:1073805. [PMID: 36861039 PMCID: PMC9968944 DOI: 10.3389/fcell.2023.1073805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/01/2023] [Indexed: 02/15/2023] Open
Abstract
Epidermal keratinocytes are enriched with at least nine connexins that are key regulators of epidermal homeostasis. The role of Cx30.3 in keratinocytes and epidermal health became evident when fourteen autosomal dominant mutations in the Cx30.3-encoding GJB4 gene were linked to a rare and incurable skin disorder called erythrokeratodermia variabilis et progressiva (EKVP). While these variants are linked to EKVP, they remain largely uncharacterized hindering therapeutic options. In this study, we characterize the expression and functional status of three EKVP-linked Cx30.3 mutants (G12D, T85P, and F189Y) in tissue-relevant and differentiation-competent rat epidermal keratinocytes. We found that GFP-tagged Cx30.3 mutants were non-functional likely due to their impaired trafficking and primary entrapment within the endoplasmic reticulum (ER). However, all mutants failed to increase BiP/GRP78 levels suggesting they were not inducing an unfolded protein response. FLAG-tagged Cx30.3 mutants were also trafficking impaired yet occasionally exhibited some capacity to assemble into gap junctions. The pathological impact of these mutants may extend beyond their trafficking deficiencies as keratinocytes expressing FLAG-tagged Cx30.3 mutants exhibited increased propidium iodide uptake in the absence of divalent cations. Attempts to rescue the delivery of trafficking impaired GFP-tagged Cx30.3 mutants into gap junctions by chemical chaperone treatment were ineffective. However, co-expression of wild type Cx30.3 greatly enhanced the assembly of Cx30.3 mutants into gap junctions, although endogenous levels of Cx30.3 do not appear to prevent the skin pathology found in patients harboring these autosomal dominant mutations. In addition, a spectrum of connexin isoforms (Cx26, Cx30, and Cx43) exhibited the differential ability to trans-dominantly rescue the assembly of GFP-tagged Cx30.3 mutants into gap junctions suggesting a broad range of connexins found in keratinocytes may favourably interact with Cx30.3 mutants. We conclude that selective upregulation of compatible wild type connexins in keratinocytes may have potential therapeutic value in rescuing epidermal defects invoked by Cx30.3 EKVP-linked mutants.
Collapse
Affiliation(s)
- Sergiu A. Lucaciu
- 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
| | - Rhett Figliuzzi
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Ruth Neumann
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Samina Nazarali
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Luigi Del Sordo
- 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
| | - Stephanie E. Leighton
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Alexandra Hauser
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Qing Shao
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Donglin Bai
- Department of Physiology and Pharmacology, University of Western Ontario, 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,*Correspondence: Dale W. Laird,
| |
Collapse
|
5
|
Interrogation of Carboxy-Terminus Localized GJA1 Variants Associated with Erythrokeratodermia Variabilis et Progressiva. Int J Mol Sci 2022; 23:ijms23010486. [PMID: 35008913 PMCID: PMC8745721 DOI: 10.3390/ijms23010486] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 02/04/2023] Open
Abstract
Although inherited GJA1 (encoding Cx43) gene mutations most often lead to oculodentodigital dysplasia and related disorders, four variants have been linked to erythrokeratodermia variabilis et progressiva (EKVP), a skin disorder characterized by erythematous and hyperkeratotic lesions. While two autosomal-dominant EKVP-linked GJA1 mutations have been shown to lead to augmented hemichannels, the consequence(s) of keratinocytes harboring a de novo P283L variant alone or in combination with a de novo T290N variant remain unknown. Interestingly, these variants reside within or adjacent to a carboxy terminus polypeptide motif that has been shown to be important in regulating the internalization and degradation of Cx43. Cx43-rich rat epidermal keratinocytes (REKs) or Cx43-ablated REKs engineered to express fluorescent protein-tagged P283L and/or T290N variants formed prototypical gap junctions at cell-cell interfaces similar to wildtype Cx43. Dye coupling and dye uptake studies further revealed that each variant or a combination of both variants formed functional gap junction channels, with no evidence of augmented hemichannel function or induction of cell death. Tracking the fate of EKVP-associated variants in the presence of the protein secretion blocker brefeldin A, or an inhibitor of protein synthesis cycloheximide, revealed that P283L or the combination of P283L and T290N variants either significantly extended Cx43 residency on the cell surface of keratinocytes or delayed its degradation. However, caution is needed in concluding that this modest change in the Cx43 life cycle is sufficient to cause EKVP, or whether an additional underlying mechanism or another unidentified gene mutation is contributing to the pathogenesis found in patients. This question will be resolved if further patients are identified where whole exome sequencing reveals a Cx43 P283L variant alone or, in combination with a T290N variant, co-segregates with EKVP across several family generations.
Collapse
|
6
|
Kordowitzki P, Sokołowska G, Wasielak-Politowska M, Skowronska A, Skowronski MT. Pannexins and Connexins: Their Relevance for Oocyte Developmental Competence. Int J Mol Sci 2021; 22:ijms22115918. [PMID: 34072911 PMCID: PMC8199496 DOI: 10.3390/ijms22115918] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/23/2021] [Accepted: 05/28/2021] [Indexed: 12/29/2022] Open
Abstract
The oocyte is the major determinant of embryo developmental competence in all mammalian species. Although fundamental advances have been generated in the field of reproductive medicine and assisted reproductive technologies in the past three decades, researchers and clinicians are still trying to elucidate molecular factors and pathways, which could be pivotal for the oocyte’s developmental competence. The cell-to-cell and cell-to-matrix communications are crucial not only for oocytes but also for multicellular organisms in general. This latter mentioned communication is among others possibly due to the Connexin and Pannexin families of large-pore forming channels. Pannexins belong to a protein group of ATP-release channels, therefore of high importance for the oocyte due to its requirements of high energy supply. An increasing body of studies on Pannexins provided evidence that these channels not only play a role during physiological processes of an oocyte but also during pathological circumstances which could lead to the development of diseases or infertility. Connexins are proteins that form membrane channels and gap-junctions, and more precisely, these proteins enable the exchange of some ions and molecules, and therefore they do play a fundamental role in the communication between the oocyte and accompanying cells. Herein, the role of Pannexins and Connexins for the processes of oogenesis, folliculogenesis, oocyte maturation and fertilization will be discussed and, at the end of this review, Pannexin and Connexin related pathologies and their impact on the developmental competence of oocytes will be provided.
Collapse
Affiliation(s)
- Paweł Kordowitzki
- Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Bydgoska Street 7, 10-243 Olsztyn, Poland;
- Department of Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Gagarina Street 7, 87-100 Torun, Poland
| | - Gabriela Sokołowska
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Jana Kilińskiego Street 1, 15-089 Białystok, Poland;
| | - Marta Wasielak-Politowska
- Center of Gynecology, Endocrinology and Reproductive Medicine—Artemida, Jagiellońska Street 78, 10-357 Olsztyn, Poland;
| | - Agnieszka Skowronska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Warszawska Street 30, 10-357 Olsztyn, Poland;
| | - Mariusz T. Skowronski
- Department of Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Gagarina Street 7, 87-100 Torun, Poland
- Correspondence: ; Tel.: +48-566-112-231
| |
Collapse
|
7
|
Gingrich J, Pu Y, Upham BL, Hulse M, Pearl S, Martin D, Avery A, Veiga-Lopez A. Bisphenol S enhances gap junction intercellular communication in ovarian theca cells. CHEMOSPHERE 2021; 263:128304. [PMID: 33155548 PMCID: PMC7726030 DOI: 10.1016/j.chemosphere.2020.128304] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 05/08/2023]
Abstract
Gap junction intercellular communication (GJIC) is necessary for ovarian function, and it is temporospatially regulated during follicular development and ovulation. At outermost layer of the antral follicle, theca cells provide structural, steroidogenic, and vascular support. Inter- and extra-thecal GJIC is required for intrafollicular trafficking of signaling molecules. Because GJIC can be altered by hormones and endocrine disrupting chemicals (EDCs), we tested if any of five common EDCs (bisphenol A (BPA), bisphenol S (BPS), bisphenol F (BPF), perfluorooctanesulfonic acid (PFOS), and triphenyltin chloride (TPT)) can interfere with theca cell GJIC. Since most chemicals are reported to repress GJIC, we hypothesized that all chemicals tested, within environmentally relevant human exposure concentrations, will inhibit theca cell GJICs. To evaluate this hypothesis, we used a scrape loading/dye transfer assay. BPS, but no other chemical tested, enhanced GJIC in a dose- and time-dependent manner in ovine primary theca cells. A signal-protein inhibitor approach was used to explore the GJIC-modulatory pathways involved. Phospholipase C and mitogen-activated protein kinase (MAPK) inhibitors significantly attenuated BPS-induced enhanced GJIC. Human theca cells were used to evaluate translational relevance of these findings. Human primary theca cells had a ∼40% increase in GJIC in response to BPS, which was attenuated with a MAPK inhibitor, suggestive of a conserved mechanism. Upregulation of GJIC could result in hyperplasia of the theca cell layer or prevent ovulation by holding the oocyte in meiotic arrest. Further studies are necessary to understand in vitro to in vivo translatability of these findings on follicle development and fertility outcomes.
Collapse
Affiliation(s)
- Jeremy Gingrich
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Yong Pu
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Brad L Upham
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI, 48824, USA
| | - Madeline Hulse
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, MI, 48912, USA
| | - Sarah Pearl
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, MI, 48912, USA
| | - Denny Martin
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, MI, 48912, USA
| | - Anita Avery
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, MI, 48912, USA; Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Almudena Veiga-Lopez
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA; Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
8
|
Boucher J, Balandre AC, Debant M, Vix J, Harnois T, Bourmeyster N, Péraudeau E, Chépied A, Clarhaut J, Debiais F, Monvoisin A, Cronier L. Cx43 Present at the Leading Edge Membrane Governs Promigratory Effects of Osteoblast-Conditioned Medium on Human Prostate Cancer Cells in the Context of Bone Metastasis. Cancers (Basel) 2020; 12:cancers12103013. [PMID: 33081404 PMCID: PMC7602984 DOI: 10.3390/cancers12103013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In its late stages, prostate cancer (PCa) is characterized by a high propensity to form osteoblastic bone metastases, mainly treated by palliative approaches. In a previous work, we demonstrated that a gap junctional protein, connexin43 (Cx43) is implicated both in the increase of aggressiveness of PCa cells and in their impact on bone. To analyze the reciprocal part of the dialogue, the current study addresses the role of Cx43 in the impact of bone microenvironment on PCa cells abilities. Using Cx43-overexpressing PCa cell lines, we determined that Cx43 is necessary for promigratory effect induced by osteoblastic conditioned media (ObCM) on individual cells. Next, we demonstrated the requirement of Cx43 membrane localization at the leading edge and the involvement of the cytoplasmic part in this ObCM-induced migration. Overall, our findings precise the role of Cx43 during PCa progression and its putative use as aggressiveness marker and as potential therapeutic targets. Abstract Among the different interacting molecules implicated in bone metastases, connexin43 (Cx43) may increase sensitivity of prostate cancer (PCa) cells to bone microenvironment, as suggested by our in silico and human tissue samples analyses that revealed increased level of Cx43 expression with PCa progression and a Cx43 specific expression in bone secondary sites. The goal of the present study was to understand how Cx43 influences PCa cells sensitivity and aggressiveness to bone microenvironment. By means of Cx43-overexpressing PCa cell lines, we revealed a Cx43-dependent promigratory effect of osteoblastic conditioned media (ObCM). This effect on directional migration relied on the presence of Cx43 at the plasma membrane and not on gap junctional intercellular communication and hemichannel functions. ObCM stimulation induced Rac1 activation and Cx43 interaction with cortactin in protrusions of migrating PCa cells. Finally, by transfecting two different truncated forms of Cx43 in LNCaP cells, we determined that the carboxy terminal (CT) part of Cx43 is crucial for the responsiveness of PCa cells to ObCM. Our study demonstrates that Cx43 level and its membrane localization modulate the phenotypic response of PCa cells to osteoblastic microenvironment and that its CT domain plays a pivotal role.
Collapse
Affiliation(s)
- Jonathan Boucher
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Annie-Claire Balandre
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Marjolaine Debant
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Justine Vix
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
- Department of Rheumatology, University Hospital Center of Poitiers, 2 Rue de la Milétrie, 86021 Poitiers, France
| | - Thomas Harnois
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Nicolas Bourmeyster
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Elodie Péraudeau
- University Hospital Center of Poitiers, 2 rue de la Milétrie, 86021 Poitiers, France; (E.P.); (J.C.)
- CNRS UMR 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), University of Poitiers, 4 Rue Michel Brunet, TSA 51106, CEDEX 09, 86073 Poitiers, France
| | - Amandine Chépied
- Laboratory of Experimental and Clinical Neurosciences, LNEC-INSERM U1084, UBM-Laboratoire de Cancérologie Biologique, CHU de Poitiers, 2 Rue de la Milétrie, 86000 Poitiers, France;
| | - Jonathan Clarhaut
- University Hospital Center of Poitiers, 2 rue de la Milétrie, 86021 Poitiers, France; (E.P.); (J.C.)
- CNRS UMR 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), University of Poitiers, 4 Rue Michel Brunet, TSA 51106, CEDEX 09, 86073 Poitiers, France
| | - Françoise Debiais
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
- Department of Rheumatology, University Hospital Center of Poitiers, 2 Rue de la Milétrie, 86021 Poitiers, France
| | - Arnaud Monvoisin
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Laurent Cronier
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
- Correspondence: ; Tel.: +33-5-49-45-37-52
| |
Collapse
|
9
|
Development of a Cx46 Targeting Strategy for Cancer Stem Cells. Cell Rep 2020; 27:1062-1072.e5. [PMID: 31018124 PMCID: PMC6497083 DOI: 10.1016/j.celrep.2019.03.079] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 01/21/2019] [Accepted: 03/21/2019] [Indexed: 12/18/2022] Open
Abstract
Gap-junction-mediated cell-cell communication enables tumor cells to synchronize complex processes. We previously found that glioblastoma cancer stem cells (CSCs) express higher levels of the gap junction protein Cx46 compared to non-stem tumor cells (non-CSCs) and that this was necessary and sufficient for CSC maintenance. To understand the mechanism underlying this requirement, we use point mutants to disrupt specific functions of Cx46 and find that Cx46-mediated gap-junction coupling is critical for CSCs. To develop a Cx46 targeting strategy, we screen a clinically relevant small molecule library and identify clofazimine as an inhibitor of Cx46-specific cell-cell communication. Clofazimine attenuates proliferation, self-renewal, and tumor growth and synergizes with temozolomide to induce apoptosis. Although clofazimine does not cross the blood-brain barrier, the combination of clofazimine derivatives optimized for brain penetrance with standard-of-care therapies may target glioblastoma CSCs. Furthermore, these results demonstrate the importance of targeting cell-cell communication as an anti-cancer therapy.
Collapse
|
10
|
Ghazizadeh Z, Kiviniemi T, Olafsson S, Plotnick D, Beerens ME, Zhang K, Gillon L, Steinbaugh MJ, Barrera V, Sui SH, Werdich AA, Kapur S, Eranti A, Gunn J, Jalkanen J, Airaksinen J, Kleber AG, Hollmén M, MacRae CA. Metastable Atrial State Underlies the Primary Genetic Substrate for MYL4 Mutation-Associated Atrial Fibrillation. Circulation 2019; 141:301-312. [PMID: 31735076 DOI: 10.1161/circulationaha.119.044268] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common clinical arrhythmia and is associated with heart failure, stroke, and increased mortality. The myocardial substrate for AF is poorly understood because of limited access to primary human tissue and mechanistic questions around existing in vitro or in vivo models. METHODS Using an MYH6:mCherry knock-in reporter line, we developed a protocol to generate and highly purify human pluripotent stem cell-derived cardiomyocytes displaying physiological and molecular characteristics of atrial cells. We modeled human MYL4 mutants, one of the few definitive genetic causes of AF. To explore non-cell-autonomous components of AF substrate, we also created a zebrafish Myl4 knockout model, which exhibited molecular, cellular, and physiologic abnormalities that parallel those in humans bearing the cognate mutations. RESULTS There was evidence of increased retinoic acid signaling in both human embryonic stem cells and zebrafish mutant models, as well as abnormal expression and localization of cytoskeletal proteins, and loss of intracellular nicotinamide adenine dinucleotide and nicotinamide adenine dinucleotide + hydrogen. To identify potentially druggable proximate mechanisms, we performed a chemical suppressor screen integrating multiple human cellular and zebrafish in vivo endpoints. This screen identified Cx43 (connexin 43) hemichannel blockade as a robust suppressor of the abnormal phenotypes in both models of MYL4 (myosin light chain 4)-related atrial cardiomyopathy. Immunofluorescence and coimmunoprecipitation studies revealed an interaction between MYL4 and Cx43 with altered localization of Cx43 hemichannels to the lateral membrane in MYL4 mutants, as well as in atrial biopsies from unselected forms of human AF. The membrane fraction from MYL4-/- human embryonic stem cell derived atrial cells demonstrated increased phospho-Cx43, which was further accentuated by retinoic acid treatment and by the presence of risk alleles at the Pitx2 locus. PKC (protein kinase C) was induced by retinoic acid, and PKC inhibition also rescued the abnormal phenotypes in the atrial cardiomyopathy models. CONCLUSIONS These data establish a mechanistic link between the transcriptional, metabolic and electrical pathways previously implicated in AF substrate and suggest novel avenues for the prevention or therapy of this common arrhythmia.
Collapse
Affiliation(s)
- Zaniar Ghazizadeh
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Tuomas Kiviniemi
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Sigurast Olafsson
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - David Plotnick
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Manu E Beerens
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kun Zhang
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Leah Gillon
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Victor Barrera
- Chan School of Public Health, Boston, MA (M.J.S., V.B., S.H.S.)
| | - Shannan Ho Sui
- Chan School of Public Health, Boston, MA (M.J.S., V.B., S.H.S.)
| | - Andreas A Werdich
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sunil Kapur
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Antti Eranti
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Jarmo Gunn
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Juho Jalkanen
- Medicity Research Laboratories (J.J., M.H.), Harvard T.H
| | - Juhani Airaksinen
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Andre G Kleber
- Department of Pathology, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, MA (A.G.K.)
| | - Maija Hollmén
- Medicity Research Laboratories (J.J., M.H.), Harvard T.H
| | - Calum A MacRae
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Genetics and Network Medicine Divisions (C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Harvard Stem Cell Institute, Boston, MA (C.A.M.)
| |
Collapse
|
11
|
Lo BKM, Sheikh S, Williams SA. In vitro and in vivo mouse follicle development in ovaries and reaggregated ovaries. Reproduction 2019; 157:135-148. [PMID: 30601757 PMCID: PMC6347279 DOI: 10.1530/rep-18-0115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 11/13/2018] [Indexed: 11/23/2022]
Abstract
Follicle development requires complex and coordinated interactions between both the oocyte and its associated somatic cells. In ovarian dysfunction, follicle development may be abnormal due to defective somatic cell function; for example, premature ovarian insufficiency or malignancies. Replacing defective somatic cells, using the reaggregated ovary (RO) technique, may 'rescue' follicle development. ROs containing mature follicles have been generated when transplanted to a host mouse to develop. We have developed a RO culture technique and the aims were to determine how follicle development differed between transplanted and cultured ROs, and the influence of ovarian age (P2 vs P6). Mouse ROs were cultured for 14 days; P2 and P6 ovaries cultured as Controls. Follicle development was compared to ROs transplanted for 14 days and ovaries from P16 and P20 mice. ROs generated from either P2 or P6 exhibited similar follicle development in culture whereas in vivo follicle development was more advanced in P6 ROs. Follicles were more developed in cultured ROs than transplanted ROs. However, follicles in cultured ROs and ovaries had smaller oocytes with fewer theca and granulosa cells than in vivo counterparts. Our results demonstrate the fluidity of follicle development despite ovary dissociation and that environment is more important to basal lamina formation and theca cell development. Furthermore, follicle development within cultured ROs appears to be independent of oocyte nest breakdown and primordial follicle formation in source ovaries. Our results highlight the need for understanding follicle development in vitro, particularly in the development of the RO technique as a potential fertility treatment.
Collapse
Affiliation(s)
- Belinda K M Lo
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Women’s Centre, Level 3, John Radcliffe Hospital, Oxford, United Kingdom
- IVF Centre, Hong Kong Sanatorium and Hospital, Happy Valley, Hong Kong
| | - Sairah Sheikh
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Women’s Centre, Level 3, John Radcliffe Hospital, Oxford, United Kingdom
| | - Suzannah A Williams
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Women’s Centre, Level 3, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
12
|
Rhett JM, Yeh ES. The Potential for Connexin Hemichannels to Drive Breast Cancer Progression through Regulation of the Inflammatory Response. Int J Mol Sci 2018; 19:ijms19041043. [PMID: 29601539 PMCID: PMC5979453 DOI: 10.3390/ijms19041043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/12/2022] Open
Abstract
Over the past few decades, connexin hemichannels have become recognized as major players in modulating the inflammatory response. Chronic inflammation is documented to promote tumorigenesis and is a critical component of tumor progression. Furthermore, inflammation is strongly linked to angiogenesis, immunotolerance, invasiveness, metastasis, and resistance in breast cancers. In this review, the literature on the role of connexin hemichannels in inflammation is summarized, and the potential role for hemichannel-mediated inflammation in driving breast cancer progression is discussed. Lastly, the potential for connexin-based therapeutics to modulate the inflammatory component of the tumor microenvironment as an avenue for the treatment of breast cancer is also discussed.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29412, USA.
| | - Elizabeth S Yeh
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29412, USA.
| |
Collapse
|
13
|
Davis HM, Aref MW, Aguilar-Perez A, Pacheco-Costa R, Allen K, Valdez S, Herrera C, Atkinson EG, Mohammad A, Lopez D, Harris MA, Harris SE, Allen M, Bellido T, Plotkin LI. Cx43 overexpression in osteocytes prevents osteocyte apoptosis and preserves cortical bone quality in aging mice. JBMR Plus 2018; 2:206-216. [PMID: 29978155 DOI: 10.1002/jbm4.10035] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Young, skeletally mature mice lacking Cx43 in osteocytes exhibit increased osteocyte apoptosis and decreased bone strength, resembling the phenotype of old mice. Further, the expression of Cx43 in bone decreases with age, suggesting a contribution of reduced Cx43 levels to the age-related changes in the skeleton. We report herein that Cx43 overexpression in osteocytes achieved by using the DMP1-8kb promoter (Cx43OT mice) attenuates the skeletal cortical, but not trabecular bone phenotype of aged, 14-month-old mice. The percentage of Cx43-expressing osteocytes was higher in Cx43OT mice, whereas the percentage of Cx43 positive osteoblasts remained similar to wild type (WT) littermate control mice. The percentage of apoptotic osteocytes and osteoblasts was increased in aged WT mice compared to skeletally mature, 6-month-old WT mice, and the percentage of apoptotic osteocytes, but not osteoblasts, was decreased in age-matched Cx43OT mice. Aged WT mice exhibited decreased bone formation and increased bone resorption as quantified by histomorphometric analysis and circulating markers, compared to skeletally mature mice. Further, aged WT mice exhibited the expected decrease in bone biomechanical structural and material properties compared to young mice. Cx43 overexpression prevented the increase in osteoclasts and decrease in bone formation on the endocortical surfaces, and the changes in circulating markers in the aged mice. Moreover, the ability of bone to resist damage was preserved in aged Cx43OT mice both at the structural and material level. All together, these findings suggest that increased Cx43 expression in osteocytes ameliorates age-induced cortical bone changes by preserving osteocyte viability and maintaining bone formation, leading to improved bone strength.
Collapse
Affiliation(s)
- Hannah M Davis
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mohammad W Aref
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexandra Aguilar-Perez
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rafael Pacheco-Costa
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kimberly Allen
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sinai Valdez
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Carmen Herrera
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Emily G Atkinson
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arwa Mohammad
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David Lopez
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Marie A Harris
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Stephen E Harris
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Matthew Allen
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Teresita Bellido
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Division of Endocrinology, Dept. Internal Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Lilian I Plotkin
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| |
Collapse
|
14
|
Rhett JM, Calder BW, Fann SA, Bainbridge H, Gourdie RG, Yost MJ. Mechanism of action of the anti-inflammatory connexin43 mimetic peptide JM2. Am J Physiol Cell Physiol 2017; 313:C314-C326. [PMID: 28701358 PMCID: PMC5625091 DOI: 10.1152/ajpcell.00229.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 12/31/2022]
Abstract
Connexin-based therapeutics have shown the potential for therapeutic efficacy in improving wound healing. Our previous work demonstrated that the connexin43 (Cx43) mimetic peptide juxtamembrane 2 (JM2) reduced the acute inflammatory response to a submuscular implant model by inhibiting purinergic signaling. Given the prospective application in improving tissue-engineered construct tolerance that these results indicated, we sought to determine the mechanism of action for JM2 in the present study. Using confocal microscopy, a gap-FRAP cell communication assay, and an ethidium bromide uptake assay of hemichannel function we found that the peptide reduced cell surface Cx43 levels, Cx43 gap junction (GJ) size, GJ communication, and hemichannel activity. JM2 is based on the sequence of the Cx43 microtubule binding domain, and microtubules have a confirmed role in intracellular trafficking of Cx43 vesicles. Therefore, we tested the effect of JM2 on Cx43-microtubule interaction and microtubule polymerization. We found that JM2 enhanced Cx43-microtubule interaction and that microtubule polymerization was significantly enhanced. Taken together, these data suggest that JM2 inhibits trafficking of Cx43 to the cell surface by promoting irrelevant microtubule polymerization and thereby reduces the number of hemichannels in the plasma membrane available to participate in proinflammatory purinergic signaling. Importantly, this work indicates that JM2 may have therapeutic value in the treatment of proliferative diseases such as cancer. We conclude that the targeted action of JM2 on Cx43 channels may improve the tolerance of implanted tissue-engineered constructs against the innate inflammatory response.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina;
| | - Bennett W Calder
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina
| | - Stephen A Fann
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina
| | - Heather Bainbridge
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, Roanoke, Virginia; and
| | - Michael J Yost
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina.,Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
15
|
Bai D, Yue B, Aoyama H. Crucial motifs and residues in the extracellular loops influence the formation and specificity of connexin docking. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:9-21. [PMID: 28693896 DOI: 10.1016/j.bbamem.2017.07.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/25/2017] [Accepted: 07/03/2017] [Indexed: 12/19/2022]
Abstract
Most of the early studies on gap junction (GJ) channel function and docking compatibility were on rodent connexins, while recent research on GJ channels gradually shifted from rodent to human connexins largely due to the fact that mutations in many human connexin genes are found to associate with inherited human diseases. The studies on human connexins have revealed some key differences from those found in rodents, calling for a comprehensive characterization of human GJ channels. Functional studies revealed that docking and formation of functional GJ channels between two hemichannels are possible only between docking-compatible connexins. Two groups of docking-compatible rodent connexins have been identified. Compatibility is believed to be due to their amino acid residue differences at the extracellular loop domains (E1 and E2). Sequence alignment of the E1 and E2 domains of all connexins known to make GJs revealed that they are highly conserved and show high sequence identity with human Cx26, which is the only connexin with near atomic resolution GJ structure. We hypothesize that different connexins have a similar structure as that of Cx26 at the E1 and E2 domains and use the corresponding residues in their E1 and E2 domains for docking. Based on the Cx26 GJ structure and sequence analysis of well-studied connexins, we propose that the E1-E1 docking interactions are staggered with each E1 interacting with two E1s on the docked connexon. The putative E1 docking residues are conserved in both docking-compatible and -incompatible connexins, indicating that E1 does not likely serve a role in docking compatibility. However, in the case of E2-E2 docking interactions, the putative docking residues are only conserved within the docking-compatible connexins, suggesting the E2 is likely to serve the function of docking compatibility. Docking compatibility studies on human connexins have attracted a lot of attention due to the fact that putative docking residues are mutational hotspots for several connexin-linked human diseases. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Donglin Bai
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.
| | - Benny Yue
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Hiroshi Aoyama
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
16
|
Roy S, Jiang JX, Li AF, Kim D. Connexin channel and its role in diabetic retinopathy. Prog Retin Eye Res 2017; 61:35-59. [PMID: 28602949 DOI: 10.1016/j.preteyeres.2017.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 05/30/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022]
Abstract
Diabetic retinopathy is the leading cause of blindness in the working age population. Unfortunately, there is no cure for this devastating ocular complication. The early stage of diabetic retinopathy is characterized by the loss of various cell types in the retina, namely endothelial cells and pericytes. As the disease progresses, vascular leakage, a clinical hallmark of diabetic retinopathy, becomes evident and may eventually lead to diabetic macular edema, the most common cause of vision loss in diabetic retinopathy. Substantial evidence indicates that the disruption of connexin-mediated cellular communication plays a critical role in the pathogenesis of diabetic retinopathy. Yet, it is unclear how altered communication via connexin channel mediated cell-to-cell and cell-to-extracellular microenvironment is linked to the development of diabetic retinopathy. Recent observations suggest the possibility that connexin hemichannels may play a role in the pathogenesis of diabetic retinopathy by allowing communication between cells and the microenvironment. Interestingly, recent studies suggest that connexin channels may be involved in regulating retinal vascular permeability. These cellular events are coordinated at least in part via connexin-mediated intercellular communication and the maintenance of retinal vascular homeostasis. This review highlights the effect of high glucose and diabetic condition on connexin channels and their impact on the development of diabetic retinopathy.
Collapse
Affiliation(s)
- Sayon Roy
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, Boston, MA, United States.
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - An-Fei Li
- Department of Ophthalmology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan
| | - Dongjoon Kim
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
17
|
Davis HM, Pacheco-Costa R, Atkinson EG, Brun LR, Gortazar AR, Harris J, Hiasa M, Bolarinwa SA, Yoneda T, Ivan M, Bruzzaniti A, Bellido T, Plotkin LI. Disruption of the Cx43/miR21 pathway leads to osteocyte apoptosis and increased osteoclastogenesis with aging. Aging Cell 2017; 16:551-563. [PMID: 28317237 PMCID: PMC5418188 DOI: 10.1111/acel.12586] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2017] [Indexed: 12/25/2022] Open
Abstract
Skeletal aging results in apoptosis of osteocytes, cells embedded in bone that control the generation/function of bone forming and resorbing cells. Aging also decreases connexin43 (Cx43) expression in bone; and osteocytic Cx43 deletion partially mimics the skeletal phenotype of old mice. Particularly, aging and Cx43 deletion increase osteocyte apoptosis, and osteoclast number and bone resorption on endocortical bone surfaces. We examined herein the molecular signaling events responsible for osteocyte apoptosis and osteoclast recruitment triggered by aging and Cx43 deficiency. Cx43‐silenced MLO‐Y4 osteocytic (Cx43def) cells undergo spontaneous cell death in culture through caspase‐3 activation and exhibit increased levels of apoptosis‐related genes, and only transfection of Cx43 constructs able to form gap junction channels reverses Cx43def cell death. Cx43def cells and bones from old mice exhibit reduced levels of the pro‐survival microRNA miR21 and, consistently, increased levels of the miR21 target phosphatase and tensin homolog (PTEN) and reduced phosphorylated Akt, whereas PTEN inhibition reduces Cx43def cell apoptosis. miR21 reduction is sufficient to induce apoptosis of Cx43‐expressing cells and miR21 deletion in miR21fl/fl bones increases apoptosis‐related gene expression, whereas a miR21 mimic prevents Cx43def cell apoptosis, demonstrating that miR21 lies downstream of Cx43. Cx43def cells release more osteoclastogenic cytokines [receptor activator of NFκB ligand (RANKL)/high‐mobility group box‐1 (HMGB1)], and caspase‐3 inhibition prevents RANKL/HMGB1 release and the increased osteoclastogenesis induced by conditioned media from Cx43def cells, which is blocked by antagonizing HMGB1‐RAGE interaction. These findings identify a novel Cx43/miR21/HMGB1/RANKL pathway involved in preventing osteocyte apoptosis that also controls osteoclast formation/recruitment and is impaired with aging.
Collapse
Affiliation(s)
- Hannah M. Davis
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Rafael Pacheco-Costa
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Emily G. Atkinson
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Lucas R. Brun
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Arancha R. Gortazar
- Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo-CEU; Madrid Spain
| | - Julia Harris
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Masahiro Hiasa
- Division of Hematology/Oncology; Department of Internal Medicine; Indiana University School of Medicine; Indianapolis IN USA
| | - Surajudeen A. Bolarinwa
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Toshiyuki Yoneda
- Division of Hematology/Oncology; Department of Internal Medicine; Indiana University School of Medicine; Indianapolis IN USA
| | - Mircea Ivan
- Division of Hematology/Oncology; Department of Internal Medicine; Indiana University School of Medicine; Indianapolis IN USA
| | - Angela Bruzzaniti
- Department of Oral Biology; Indiana University School of Dentistry; Indianapolis IN USA
| | - Teresita Bellido
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
- Division of Endocrinology; Department of Internal Medicine; Indiana University School of Medicine; Indianapolis IN USA
- Roudebush Veterans Administration Medical Center; Indianapolis IN USA
| | - Lilian I. Plotkin
- Department of Anatomy & Cell Biology; Indiana University School of Medicine; Indianapolis IN USA
- Roudebush Veterans Administration Medical Center; Indianapolis IN USA
| |
Collapse
|
18
|
Gajardo-Gómez R, Labra VC, Orellana JA. Connexins and Pannexins: New Insights into Microglial Functions and Dysfunctions. Front Mol Neurosci 2016; 9:86. [PMID: 27713688 PMCID: PMC5031785 DOI: 10.3389/fnmol.2016.00086] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/05/2016] [Indexed: 12/11/2022] Open
Abstract
Under physiological conditions, microglia adopt a resting phenotype associated with the production of anti-inflammatory and neurotrophic factors. In response to a wide variety of insults, these cells shift to an activated phenotype that is necessary for the proper restoration of brain homeostasis. However, when the intensity of a threat is relatively high, microglial activation worsens the progression of damage rather than providing protection, with potentially significant consequences for neuronal survival. Coordinated interactions among microglia and other brain cells, including astrocytes and neurons, are critical for the development of timely and optimal inflammatory responses in the brain parenchyma. Tissue synchronization is in part mediated by connexins and pannexins, which are protein families that form different plasma membrane channels to communicate with neighboring cells. Gap junction channels (which are exclusively formed by connexins in vertebrates) connect the cytoplasm of contacting cells to coordinate electrical and metabolic coupling. Hemichannels (HCs) and pannexons (which are formed by connexins and pannexins, respectively) communicate the intra- and extracellular compartments and serve as diffusion pathways for the exchange of ions and small molecules. In this review article, we discuss the available evidence concerning the functional expression and regulation of connexin- and pannexin-based channels in microglia and their contributions to microglial function and dysfunction. Specifically, we focus on the possible implications of these channels in microglia-to-microglia, microglia-to-astrocyte and neuron-to-microglia interactions in the inflamed brain.
Collapse
Affiliation(s)
- Rosario Gajardo-Gómez
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Valeria C Labra
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
19
|
Kelly JJ, Esseltine JL, Shao Q, Jabs EW, Sampson J, Auranen M, Bai D, Laird DW. Specific functional pathologies of Cx43 mutations associated with oculodentodigital dysplasia. Mol Biol Cell 2016; 27:2172-85. [PMID: 27226478 PMCID: PMC4945137 DOI: 10.1091/mbc.e16-01-0062] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/20/2016] [Indexed: 02/02/2023] Open
Abstract
Oculodentodigital dysplasia (ODDD) is a rare genetic disease that affects the development of multiple organs in the human body. More than 70 mutations in the gap junction connexin43 (Cx43) gene, GJA1, are associated with ODDD, most of which are inherited in an autosomal dominant manner. Many patients exhibit similar clinical presentations. However, there is high intrafamilial and interfamilial phenotypic variability. To better understand this variability, we established primary human dermal fibroblast cultures from several ODDD patients and unaffected controls. In the present study, we characterized three fibroblast lines expressing heterozygous p.L7V, p.G138R, and p.G143S Cx43 variants. All ODDD fibroblasts exhibited slower growth, reduced migration, and defective cell polarization, traits common to all ODDD fibroblasts studied so far. However, we found striking differences in overall expression levels, with p.L7V down-regulated at the mRNA and protein level. Although all of the Cx43 variants could traffic to the cell surface, there were stark differences in gap junction plaque formation, gap junctional intercellular communication, Cx43 phosphorylation, and hemichannel activity among Cx43 variants, as well as subtle differences in myofibroblast differentiation. Together these findings enabled us to discover mutation-specific pathologies that may help to predict future clinical outcomes.
Collapse
Affiliation(s)
- John J Kelly
- Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Jessica L Esseltine
- Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Qing Shao
- Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Ethylin Wang Jabs
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Johns Hopkins University, Baltimore, MD 21205
| | - Jacinda Sampson
- Department of Neurology, Stanford University Medical Center, Palo Alto, CA 94304
| | - Mari Auranen
- Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital, Helsinki 00290, Finland
| | - Donglin Bai
- Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Dale W Laird
- Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
20
|
Loss of Panx1 Impairs Mammary Gland Development at Lactation: Implications for Breast Tumorigenesis. PLoS One 2016; 11:e0154162. [PMID: 27099931 PMCID: PMC4839729 DOI: 10.1371/journal.pone.0154162] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/08/2016] [Indexed: 12/03/2022] Open
Abstract
Pannexin1 (Panx1) subunits oligomerize to form large-pore channels between the intracellular and extracellular milieu that have been shown to regulate proliferation, differentiation and cell death mechanisms. These key cellular responses are ultimately necessary for normal tissue development and function but the role of Panx1 in development, differentiation and function in many tissues remains unexplored, including that of the breast. Panx1 was identified to be expressed in the mammary gland through western blot and immunofluorescent analysis and is dynamically upregulated during pregnancy and lactation. In order to evaluate the role of Panx1 in the context of mammary gland development and function, Panx1-/- mice were evaluated in comparison to wild-type mice in the mammary glands of virgin, lactating and involuting mice. Our results revealed that Panx1 ablation did not affect virgin or involuting mammary glands following histological and whole mount analysis. Panx1 was necessary for timely alveolar development during early lactation based on a decreased number of alveolar lumen following histological analysis and reduced proliferation following Ki67 immunofluorescent labelling. Importantly, the loss of Panx1 in lactating mammary glands did not overtly affect epithelial or secretory differentiation of the mammary gland suggesting that Panx1 is not critical in normal mammary gland function. In addition, PANX1 mRNA expression was correlated with negative clinical outcomes in patients with breast cancer using in silico arrays. Together, our results suggest that Panx1 is necessary for timely alveolar development following the transition from pregnancy to lactation, which may have implications extending to patients with breast cancer.
Collapse
|
21
|
Laird DW, Naus CC. Gerald M. Kidder - bridging the gap in cell and developmental biology. Semin Cell Dev Biol 2016; 50:1-3. [PMID: 26940063 DOI: 10.1016/j.semcdb.2016.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Christian C Naus
- Department of Cellular & Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
22
|
Kibschull M, Gellhaus A, Carette D, Segretain D, Pointis G, Gilleron J. Physiological roles of connexins and pannexins in reproductive organs. Cell Mol Life Sci 2015; 72:2879-98. [PMID: 26100514 PMCID: PMC11114083 DOI: 10.1007/s00018-015-1965-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/11/2015] [Indexed: 12/15/2022]
Abstract
Reproductive organs are complex and well-structured tissues essential to perpetuate the species. In mammals, the male and female reproductive organs vary on their organization, morphology and function. Connectivity between cells in such tissues plays pivotal roles in organogenesis and tissue functions through the regulation of cellular proliferation, migration, differentiation and apoptosis. Connexins and pannexins can be seen as major regulators of these physiological processes. In the present review, we assembled several lines of evidence demonstrating that these two families of proteins are essential for male and female reproduction.
Collapse
Affiliation(s)
- Mark Kibschull
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Toronto, M5T 3H7 Canada
| | - Alexandra Gellhaus
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Diane Carette
- UMR S1147, University Paris Descartes, 45 rue des Saints-Pères, 75006 Paris, France
- University of Versailles, 78035 Saint Quentin, France
- INSERM U 1065, University of Nice Sophia-Antipolis, 151 Route Saint-Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex 3, France
| | - Dominique Segretain
- UMR S1147, University Paris Descartes, 45 rue des Saints-Pères, 75006 Paris, France
- University of Versailles, 78035 Saint Quentin, France
| | - Georges Pointis
- INSERM U 1065, University of Nice Sophia-Antipolis, 151 Route Saint-Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex 3, France
| | - Jerome Gilleron
- INSERM U 1065, University of Nice Sophia-Antipolis, 151 Route Saint-Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex 3, France
| |
Collapse
|
23
|
Abstract
Adenosine-5'-triphosphate is released by neuroendocrine, endocrine, and other cell types and acts as an extracellular agonist for ligand-gated P2X cationic channels and G protein-coupled P2Y receptors in numerous organs and tissues, including the endocrine system. The breakdown of ATP by ectonucleotidases not only terminates its extracellular messenger functions, but also provides a pathway for the generation of two additional agonists: adenosine 5'-diphosphate, acting via some P2Y receptors, and adenosine, a native agonist for G protein-coupled adenosine receptors, also expressed in the endocrine system. This article provides a review of purinergic signaling pathways in the hypothalamic magnocellular neurosecretory cells and neurohypophysis, hypothalamic parvocellular neuroendocrine system, adenohypophysis, and effector glands organized in five axes: hypothalamic-pituitary-gonadal, hypothalamic-pituitary-thyroid, hypothalamic-pituitary-adrenal, hypothalamic-pituitary-growth hormone, and hypothalamic-pituitary-prolactin. We attempted to summarize current knowledge of purinergic receptor subtypes expressed in the endocrine system, including their roles in intracellular signaling, hormone secretion, and other cell functions. We also briefly review the release mechanism for adenosine-5'-triphosphate by neuroendocrine, endocrine and surrounding cells, the enzymes involved in adenosine-5'-triphosphate hydrolysis to adenosine-5'-diphosphate and adenosine, and the relevance of this pathway for sequential activation of receptors and termination of signaling.
Collapse
|
24
|
Winterhager E, Kidder GM. Gap junction connexins in female reproductive organs: implications for women's reproductive health. Hum Reprod Update 2015; 21:340-52. [PMID: 25667189 DOI: 10.1093/humupd/dmv007] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/20/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Connexins comprise a family of ~20 proteins that form intercellular membrane channels (gap junction channels) providing a direct route for metabolites and signalling molecules to pass between cells. This review provides a critical analysis of the evidence for essential roles of individual connexins in female reproductive function, highlighting implications for women's reproductive health. METHODS No systematic review has been carried out. Published literature from the past 35 years was surveyed for research related to connexin involvement in development and function of the female reproductive system. Because of the demonstrated utility of genetic manipulation for elucidating connexin functions in various organs, much of the cited information comes from research with genetically modified mice. In some cases, a distinction is drawn between connexin functions clearly related to the formation of gap junction channels and those possibly linked to non-channel roles. RESULTS AND CONCLUSIONS Based on work with mice, several connexins are known to be required for female reproductive functions. Loss of connexin43 (CX43) causes an oocyte deficiency, and follicles lacking or expressing less CX43 in granulosa cells exhibit reduced growth, impairing fertility. CX43 is also expressed in human cumulus cells and, in the context of IVF, has been correlated with pregnancy outcome, suggesting that this connexin may be a determinant of oocyte and embryo quality in women. Loss of CX37, which exclusively connects oocytes with granulosa cells in the mouse, caused oocytes to cease growing without acquiring meiotic competence. Blocking of CX26 channels in the uterine epithelium disrupted implantation whereas loss or reduction of CX43 expression in the uterine stroma impaired decidualization and vascularization in mouse and human. Several connexins are important in placentation and, in the human, CX43 is a key regulator of the fusogenic pathway from the cytotrophoblast to the syncytiotrophoblast, ensuring placental growth. CX40, which characterizes the extravillous trophoblast (EVT), supports proliferation of the proximal EVTs while preventing them from differentiating into the invasive pathway. Furthermore, women with recurrent early pregnancy loss as well as those with endometriosis exhibit reduced levels of CX43 in their decidua. The antimalaria drug mefloquine, which blocks gap junction function, is responsible for increased risk of early pregnancy loss and stillbirth, probably due to inhibition of intercellular communication in the decidua or between trophoblast layers followed by an impairment of placental growth. Gap junctions also play a critical role in regulating uterine blood flow, contributing to the adaptive response to pregnancy. Given that reproductive impairment can result from connexin mutations in mice, it is advised that women suffering from somatic disease symptoms associated with connexin gene mutations be additionally tested for impacts on reproductive function. Better knowledge of these essential connexin functions in human female reproductive organs is important for safeguarding women's reproductive health.
Collapse
Affiliation(s)
- Elke Winterhager
- Institute of Molecular Biology, University of Duisburg-Essen, University Clinics, 45211 Essen, Germany
| | - Gerald M Kidder
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario and Children's Health Research Institute, London, Ontario N6C 2V5, Canada
| |
Collapse
|
25
|
Good ME, Ek-Vitorín JF, Burt JM. Structural determinants and proliferative consequences of connexin 37 hemichannel function in insulinoma cells. J Biol Chem 2014; 289:30379-30386. [PMID: 25217644 PMCID: PMC4215222 DOI: 10.1074/jbc.m114.583054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 09/10/2014] [Indexed: 01/24/2023] Open
Abstract
Connexin (Cx) 37 suppresses vascular and cancer cell proliferation. The C terminus and a channel able to function are necessary, and neither by itself is sufficient, for Cx37 to mediate growth suppression. Cx37 supports transmembrane and intercellular signaling by forming functional hemichannels (HCs) and gap junction channels (GJCs), respectively. Here we determined whether Cx37 with HC, but not GJC, functionality would suppress proliferation of rat insulinoma (Rin) cells comparably to wild-type Cx37 (Cx37-WT). We mutated extracellular loop residues hypothesized to compromise HC docking but not HC function (six cysteines mutated to alanine, C54A,C61A,C65A, C187A,C192A,C198A (designated as C6A); N55I; and Q58L). All three mutants trafficked to the plasma membrane and formed protein plaques comparably to Cx37-WT. None of the mutants formed functional GJCs, and Cx37-C6A did not form functional HCs. Cx37-N55I and -Q58L formed HCs with behavior and permeation properties similar to Cx37-WT (especially Q58L), but none of the mutants suppressed Rin cell proliferation. The data indicate that determinants of Cx37 HC function differ from other Cxs and that HC functions with associated HC-supported protein-protein interactions are not sufficient for Cx37 to suppress Rin cell proliferation. Together with previously published data, these results suggest that Cx37 suppresses Rin cell proliferation only when in a specific conformation achieved by interaction of the C terminus with a Cx37 pore-forming domain able to open as a GJC.
Collapse
Affiliation(s)
- Miranda E Good
- Department of Physiology, University of Arizona, Tucson, Arizona 85724-5051
| | - José F Ek-Vitorín
- Department of Physiology, University of Arizona, Tucson, Arizona 85724-5051
| | - Janis M Burt
- Department of Physiology, University of Arizona, Tucson, Arizona 85724-5051.
| |
Collapse
|
26
|
Berger AC, Kelly JJ, Lajoie P, Shao Q, Laird DW. Mutations in Cx30 that are linked to skin disease and non-syndromic hearing loss exhibit several distinct cellular pathologies. J Cell Sci 2014; 127:1751-64. [DOI: 10.1242/jcs.138230] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
ABSTRACT
Connexin 30 (Cx30), a member of the large gap-junction protein family, plays a role in the homeostasis of the epidermis and inner ear through gap junctional intercellular communication (GJIC). Here, we investigate the underlying mechanisms of four autosomal dominant Cx30 gene mutations that are linked to hearing loss and/or various skin diseases. First, the T5M mutant linked to non-syndromic hearing loss formed functional gap junction channels and hemichannels, similar to wild-type Cx30. The loss-of-function V37E mutant associated with Clouston syndrome or keratitis-ichthyosis-deafness syndrome was retained in the endoplasmic reticulum and significantly induced apoptosis. The G59R mutant linked to the Vohwinkel and Bart-Pumphrey syndromes was retained primarily in the Golgi apparatus and exhibited loss of gap junction channel and hemichannel function but did not cause cell death. Lastly, the A88V mutant, which is linked to the development of Clouston syndrome, also significantly induced apoptosis but through an endoplasmic-reticulum-independent mechanism. Collectively, we discovered that four unique Cx30 mutants might cause disease through different mechanisms that also likely include their selective trans-dominant effects on coexpressed connexins, highlighting the overall complexity of connexin-linked diseases and the importance of GJIC in disease prevention.
Collapse
Affiliation(s)
- Amy C. Berger
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - John J. Kelly
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Patrick Lajoie
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Qing Shao
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Dale W. Laird
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
27
|
Syndromic and non-syndromic disease-linked Cx43 mutations. FEBS Lett 2014; 588:1339-48. [PMID: 24434540 DOI: 10.1016/j.febslet.2013.12.022] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 12/30/2013] [Indexed: 01/05/2023]
Abstract
There are now at least 14 distinct diseases linked to germ line mutations in the 21 genes that encode the connexin (Cx) family of gap junction proteins. This review focuses on the links between germ-line mutations in the gene encoding Cx43 (GJA1) and the human disease termed oculodentodigital dysplasia (ODDD). This disease is clinically characterized by soft tissue fusion of the digits, abnormal craniofacial bone development, small eyes and loss of tooth enamel. However, the disease is considerably more complex and somewhat degenerative as patients often suffer from other syndromic effects that include incontinence, glaucoma, skin diseases and neuropathies that become more pronounced during aging. The challenge continues to be understanding how distinct Cx43 gene mutations cause such a diverse range of tissue phenotypes and pathophysiological changes while other Cx43-rich organs are relatively unaffected. This review will provide an overview of many of these studies and distill some themes and outstanding questions that need to be addressed in the coming years.
Collapse
|
28
|
Nualart-Marti A, del Molino EM, Grandes X, Bahima L, Martin-Satué M, Puchal R, Fasciani I, González-Nieto D, Ziganshin B, Llobet A, Barrio LC, Solsona C. Role of connexin 32 hemichannels in the release of ATP from peripheral nerves. Glia 2013; 61:1976-89. [PMID: 24123415 DOI: 10.1002/glia.22568] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 11/12/2022]
Abstract
Extracellular purines elicit strong signals in the nervous system. Adenosine-5'-triphosphate (ATP) does not spontaneously cross the plasma membrane, and nervous cells secrete ATP by exocytosis or through plasma membrane proteins such as connexin hemichannels. Using a combination of imaging, luminescence and electrophysiological techniques, we explored the possibility that Connexin 32 (Cx32), expressed in Schwann cells (SCs) myelinating the peripheral nervous system could be an important source of ATP in peripheral nerves. We triggered the release of ATP in vivo from mice sciatic nerves by electrical stimulation and from cultured SCs by high extracellular potassium concentration-evoked depolarization. No ATP was detected in the extracellular media after treatment of the sciatic nerve with Octanol or Carbenoxolone, and ATP release was significantly inhibited after silencing Cx32 from SCs cultures. We investigated the permeability of Cx32 to ATP by expressing Cx32 hemichannels in Xenopus laevis oocytes. We found that ATP release is coupled to the inward tail current generated after the activation of Cx32 hemichannels by depolarization pulses, and it is sensitive to low extracellular calcium concentrations. Moreover, we found altered ATP release in mutated Cx32 hemichannels related to the X-linked form of Charcot-Marie-Tooth disease, suggesting that purinergic-mediated signaling in peripheral nerves could underlie the physiopathology of this neuropathy.
Collapse
Affiliation(s)
- Anna Nualart-Marti
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine-Campus Bellvitge, University of Barcelona, Hospitalet del Llobregat, Barcelona, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet del Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
De Bock M, Kerrebrouck M, Wang N, Leybaert L. Neurological manifestations of oculodentodigital dysplasia: a Cx43 channelopathy of the central nervous system? Front Pharmacol 2013; 4:120. [PMID: 24133447 PMCID: PMC3783840 DOI: 10.3389/fphar.2013.00120] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 09/02/2013] [Indexed: 12/30/2022] Open
Abstract
The coordination of tissue function is mediated by gap junctions (GJs) that enable direct cell–cell transfer of metabolic and electric signals. GJs are formed by connexins of which Cx43 is most widespread in the human body. In the brain, Cx43 GJs are mostly found in astroglia where they coordinate the propagation of Ca2+ waves, spatial K+ buffering, and distribution of glucose. Beyond its role in direct intercellular communication, Cx43 also forms unapposed, non-junctional hemichannels in the plasma membrane of glial cells. These allow the passage of several neuro- and gliotransmitters that may, combined with downstream paracrine signaling, complement direct GJ communication among glial cells and sustain glial-neuronal signaling. Mutations in the GJA1 gene encoding Cx43 have been identified in a rare, mostly autosomal dominant syndrome called oculodentodigital dysplasia (ODDD). ODDD patients display a pleiotropic phenotype reflected by eye, hand, teeth, and foot abnormalities, as well as craniofacial and bone malformations. Remarkably, neurological symptoms such as dysarthria, neurogenic bladder (manifested as urinary incontinence), spasticity or muscle weakness, ataxia, and epilepsy are other prominent features observed in ODDD patients. Over 10 mutations detected in patients diagnosed with neurological disorders are associated with altered functionality of Cx43 GJs/hemichannels, but the link between ODDD-related abnormal channel activities and neurologic phenotype is still elusive. Here, we present an overview on the nature of the mutants conveying structural and functional changes of Cx43 channels and discuss available evidence for aberrant Cx43 GJ and hemichannel function. In a final step, we examine the possibilities of how channel dysfunction may lead to some of the neurological manifestations of ODDD.
Collapse
Affiliation(s)
- Marijke De Bock
- Physiology Group, Department of Basic Medical Sciences, Ghent University Ghent, Belgium
| | | | | | | |
Collapse
|
30
|
Stojilkovic SS, Zemkova H. P2X receptor channels in endocrine glands. WILEY INTERDISCIPLINARY REVIEWS. MEMBRANE TRANSPORT AND SIGNALING 2013; 2:173-180. [PMID: 24073387 PMCID: PMC3780426 DOI: 10.1002/wmts.89] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The endocrine system is the system of ductless glands and single cells that synthetize hormones and release them directly into the bloodstream. Regulation of endocrine system is very complex and ATP and its degradable products ADP and adenosine contribute to its regulation acting as extracellular messengers for purinergic receptors. These include P2X receptors, a family of ligand-gated ion channels which expression and roles in endocrine tissues are reviewed here. There are seven mammalian purinergic receptor subunits, denoted P2X1 through P2X7, and the majority of these subunits are also expressed in secretory and non-secretory cells of endocrine system. Functional channels have been identified in the neuroendocrine hypothalamus, the posterior and anterior pituitary, the thyroid gland, the adrenals, the endocrine pancreas, the gonads and the placenta. Native channels are capable of promoting calcium influx through its pore in both excitable and non-excitable cells, as well as of increasing electrical activity in excitable cells by membrane depolarization. This leads to generation of calcium transients and stimulation of hormone release. The pattern of expression and action of P2XRs in endocrine system suggests that locally produced ATP amplifies and synchronizes the secretory responses of individual cells.
Collapse
Affiliation(s)
- Stanko S. Stojilkovic
- Section on Cellular Signaling, Program in Developmental Neuroscience, The Eunice Kennedy Shiver National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-4510
| | - Hana Zemkova
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology of the Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
31
|
Plotkin LI, Bellido T. Beyond gap junctions: Connexin43 and bone cell signaling. Bone 2013; 52:157-66. [PMID: 23041511 PMCID: PMC3513515 DOI: 10.1016/j.bone.2012.09.030] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 09/20/2012] [Accepted: 09/25/2012] [Indexed: 12/31/2022]
Abstract
Connexin43 (Cx43) is the most abundant gap junction protein expressed in bone cells and plays a central role in cell-to-cell communication in the skeleton. Findings of the last decade uncovered functions of Cx43 hemichannels expressed on unopposed plasma cell membranes as mediators of the communication between bone cells and their extracellular milieu. Additionally, through its cytoplasmic C-terminus domain, Cx43 serves as a scaffolding protein that associates with structural and signaling molecules leading to regulation of intracellular signaling, independent of channel activity. This perspective discusses the evidence demonstrating that via these diverse mechanisms Cx43 is a key component of the intracellular machinery responsible for signal transduction in bone in response to pharmacologic, hormonal and mechanical stimuli. This advance in the knowledge of the role of connexins increases our understanding of the pathophysiological mechanisms that regulate bone cell function and provides new opportunities to treat bone diseases.
Collapse
Affiliation(s)
- Lilian I. Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
32
|
Kar R, Batra N, Riquelme MA, Jiang JX. Biological role of connexin intercellular channels and hemichannels. Arch Biochem Biophys 2012; 524:2-15. [PMID: 22430362 PMCID: PMC3376239 DOI: 10.1016/j.abb.2012.03.008] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 02/16/2012] [Accepted: 03/06/2012] [Indexed: 12/11/2022]
Abstract
Gap junctions (GJ) and hemichannels (HC) formed from the protein subunits called connexins are transmembrane conduits for the exchange of small molecules and ions. Connexins and another group of HC-forming proteins, pannexins comprise the two families of transmembrane proteins ubiquitously distributed in vertebrates. Most cell types express more than one connexin or pannexin. While connexin expression and channel activity may vary as a function of physiological and pathological states of the cell and tissue, only a few studies suggest the involvement of pannexin HC in acquired pathological conditions. Importantly, genetic mutations in connexin appear to interfere with GJ and HC function which results in several diseases. Thus connexins could serve as potential drug target for therapeutic intervention. Growing evidence suggests that diseases resulting from HC dysfunction might open a new direction for development of specific HC reagents. This review provides a comprehensive overview of the current studies of GJ and HC formed by connexins and pannexins in various tissue and organ systems including heart, central nervous system, kidney, mammary glands, ovary, testis, lens, retina, inner ear, bone, cartilage, lung and liver. In addition, present knowledge of the role of GJ and HC in cell cycle progression, carcinogenesis and stem cell development is also discussed.
Collapse
Affiliation(s)
| | | | - Manuel A Riquelme
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900
| | - Jean X. Jiang
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900
| |
Collapse
|
33
|
Wang N, De Bock M, Decrock E, Bol M, Gadicherla A, Vinken M, Rogiers V, Bukauskas FF, Bultynck G, Leybaert L. Paracrine signaling through plasma membrane hemichannels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:35-50. [PMID: 22796188 DOI: 10.1016/j.bbamem.2012.07.002] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 06/29/2012] [Accepted: 07/06/2012] [Indexed: 12/24/2022]
Abstract
Plasma membrane hemichannels composed of connexin (Cx) proteins are essential components of gap junction channels but accumulating evidence suggests functions of hemichannels beyond the communication provided by junctional channels. Hemichannels not incorporated into gap junctions, called unapposed hemichannels, can open in response to a variety of signals, electrical and chemical, thereby forming a conduit between the cell's interior and the extracellular milieu. Open hemichannels allow the bidirectional passage of ions and small metabolic or signaling molecules of below 1-2kDa molecular weight. In addition to connexins, hemichannels can also be formed by pannexin (Panx) proteins and current evidence suggests that Cx26, Cx32, Cx36, Cx43 and Panx1, form hemichannels that allow the diffusive release of paracrine messengers. In particular, the case is strong for ATP but substantial evidence is also available for other messengers like glutamate and prostaglandins or metabolic substances like NAD(+) or glutathione. While this field is clearly in expansion, evidence is still lacking at essential points of the paracrine signaling cascade that includes not only messenger release, but also downstream receptor signaling and consequent functional effects. The data available at this moment largely derives from in vitro experiments and still suffers from the difficulty of separating the functions of connexin-based hemichannels from gap junctions and from pannexin hemichannels. However, messengers like ATP or glutamate have universal roles in the body and further defining the contribution of hemichannels as a possible release pathway is expected to open novel avenues for better understanding their contribution to a variety of physiological and pathological processes. This article is part of a Special Issue entitled: The Communicating junctions, roles and dysfunctions.
Collapse
Affiliation(s)
- Nan Wang
- Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Good ME, Ek-Vitorín JF, Burt JM. Extracellular loop cysteine mutant of cx37 fails to suppress proliferation of rat insulinoma cells. J Membr Biol 2012; 245:369-80. [PMID: 22797939 PMCID: PMC3527626 DOI: 10.1007/s00232-012-9459-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 06/20/2012] [Indexed: 11/29/2022]
Abstract
Although a functional pore domain is required for connexin 37 (Cx37)-mediated suppression of rat insulinoma (Rin) cell proliferation, it is unknown whether functional hemichannels would be sufficient or if Cx37 gap junction channels are required for growth suppression. To test this possibility, we targeted extracellular loop cysteines for mutation, expecting that the mutated protein would retain hemichannel, but not gap junction channel, functionality. Cysteines at positions 61 and 65 in the first extracellular loop of Cx37 were mutated to alanine and the mutant protein (Cx37-C61,65A) expressed in Rin cells. Although the resulting iRin37-C61,65A cells expressed the mutant protein comparably to Cx37 wild-type (Cx37-WT)--expressing Rin cells (iRin37), Cx37-C61,65A expression did not suppress the proliferation of Rin cells. As expected, iRin37-C61,65A cells did not form functional gap junction channels. However, functional hemichannels also could not be detected in iRin37-C61,65A cells by either dye uptake or electrophysiological approaches. Thus, failure of Cx37-C61,65A to suppress the proliferation of Rin cells is consistent with previous data demonstrating the importance of channel functionality to Cx37's growth-suppressive function. Moreover, failure of the Cx37-C61,65A hemichannel to function, even in low external calcium, emphasizes the importance of extracellular loop cysteines not only in hemichannel docking but also in determining the ability of the hemichannel to adopt a closed configuration that can open in response to triggers, such as low external calcium, effective at opening Cx37-WT hemichannels.
Collapse
Affiliation(s)
- Miranda E. Good
- Department of Physiology, University of Arizona, PO Box 245051, Tucson, AZ 85724, USA
| | - José F. Ek-Vitorín
- Department of Physiology, University of Arizona, PO Box 245051, Tucson, AZ 85724, USA
| | - Janis M. Burt
- Department of Physiology, University of Arizona, PO Box 245051, Tucson, AZ 85724, USA
| |
Collapse
|
35
|
Dyce PW, Norris RP, Lampe PD, Kidder GM. Phosphorylation of serine residues in the C-terminal cytoplasmic tail of connexin43 regulates proliferation of ovarian granulosa cells. J Membr Biol 2012; 245:291-301. [PMID: 22729691 DOI: 10.1007/s00232-012-9450-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 06/01/2012] [Indexed: 10/28/2022]
Abstract
Connexin43 (Cx43) forms gap junctions that couple the granulosa cells of ovarian follicles. In Cx43 knockout mice, follicle growth is restricted as a result of impaired granulosa cell proliferation. We have used these mice to examine the importance of specific Cx43 phosphorylation sites in follicle growth. Serines at residues 255, 262, 279, and 282 are MAP kinase substrates that, when phosphorylated, reduce junctional conductance. Mutant forms of Cx43 were constructed with these serines replaced with amino acids that cannot be phosphorylated. These mutants were transduced into Cx43 knockout ovarian somatic cells that were combined with wild-type oocytes and grafted into immunocompromised female mice permitting follicle growth in vivo. Despite residues 255 or 262 being mutated to prevent their being phosphorylated, recombinant ovaries constructed with these mutants were able to rescue the null phenotype, restoring complete folliculogenesis. In contrast, Cx43 with serine to alanine mutations at both residues 279 and 282 or at all four residues failed to rescue folliculogenesis; the mutant molecules were largely confined to intracellular sites, with few gap junctions. Using an in vitro proliferation assay, we confirmed a decrease in proliferation of granulosa cells expressing the double mutant construct. These results indicate that Cx43 phosphorylation by MAP kinase at serines 279 and 282 occurs in granulosa cells of early follicles and that this is involved in regulating follicle development.
Collapse
Affiliation(s)
- Paul W Dyce
- Department of Physiology and Pharmacology, The University of Western Ontario and Children's Health Research Institute, London, ON N6C 2V5, Canada
| | | | | | | |
Collapse
|
36
|
Bivi N, Lezcano V, Romanello M, Bellido T, Plotkin LI. Connexin43 interacts with βarrestin: a pre-requisite for osteoblast survival induced by parathyroid hormone. J Cell Biochem 2012; 112:2920-30. [PMID: 21630325 DOI: 10.1002/jcb.23208] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Parathyroid hormone (PTH) promotes osteoblast survival through a mechanism that depends on cAMP-mediated signaling downstream of the G protein-coupled receptor PTHR1. We present evidence herein that PTH-induced survival signaling is impaired in cells lacking connexin43 (Cx43). Thus, expression of functional Cx43 dominant negative proteins or Cx43 knock-down abolished the expression of cAMP-target genes and anti-apoptosis induced by PTH in osteoblastic cells. In contrast, cells lacking Cx43 were still responsive to the stable cAMP analog dibutyril-cAMP. PTH survival signaling was rescued by transfecting wild type Cx43 or a truncated dominant negative mutant of βarrestin, a PTHR1-interacting molecule that limits cAMP signaling. On the other hand, Cx43 mutants lacking the cytoplasmic domain (Cx43(Δ245)) or unable to be phosphorylated at serine 368 (Cx43(S368A)), a residue crucial for Cx43 trafficking and function, failed to restore the anti-apoptotic effect of PTH in Cx43-deficient cells. In addition, overexpression of wild type βarrestin abrogated PTH survival signaling in Cx43-expressing cells. Moreover, βarrestin physically associated in vivo to wild type Cx43 and to a lesser extent to Cx43(S368A) ; and this association and the phosphorylation of Cx43 in serine 368 were reduced by PTH. Furthermore, induction of Cx43(S368) phosphorylation or overexpression of wild type Cx43, but not Cx43(Δ245) or Cx43(S368A) , reduced the interaction between βarrestin and the PTHR1. These studies demonstrate that βarrestin is a novel Cx43-interacting protein and suggest that, by sequestering βarrestin, Cx43 facilitates cAMP signaling, thereby exerting a permissive role on osteoblast survival induced by PTH.
Collapse
Affiliation(s)
- Nicoletta Bivi
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202-5120, USA
| | | | | | | | | |
Collapse
|
37
|
Abstract
The appearance of multicellular organisms imposed the development of several mechanisms for cell-to-cell communication, whereby different types of cells coordinate their function. Some of these mechanisms depend on the intercellular diffusion of signal molecules in the extracellular spaces, whereas others require cell-to-cell contact. Among the latter mechanisms, those provided by the proteins of the connexin family are widespread in most tissues. Connexin signaling is achieved via direct exchanges of cytosolic molecules between adjacent cells at gap junctions, for cell-to-cell coupling, and possibly also involves the formation of membrane "hemi-channels," for the extracellular release of cytosolic signals, direct interactions between connexins and other cell proteins, and coordinated influence on the expression of multiple genes. Connexin signaling appears to be an obligatory attribute of all multicellular exocrine and endocrine glands. Specifically, the experimental evidence we review here points to a direct participation of the Cx36 isoform in the function of the insulin-producing β-cells of the endocrine pancreas, and of the Cx40 isoform in the function of the renin-producing juxtaglomerular epithelioid cells of the kidney cortex.
Collapse
Affiliation(s)
- Domenico Bosco
- Department of Surgery, University of Geneva Medical School, Geneva, Switzerland
| | | | | |
Collapse
|
38
|
Scemes E. Nature of plasmalemmal functional "hemichannels". BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:1880-3. [PMID: 21703226 DOI: 10.1016/j.bbamem.2011.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 06/04/2011] [Accepted: 06/07/2011] [Indexed: 12/28/2022]
Abstract
The molecular identity of the protein forming "hemichannels" at non-junctional membranes is disputed. The family of gap junction proteins, innexins, connexins, and pannexins share several common features, including permeability characteristics and sensitivity to blocking agents. Such overlap in properties renders the identification of which of these protein species actually establishes the non-junctional membrane conductance and permeability quite complicated, especially because in vertebrates pannexins and connexins have largely overlapping distributions in tissues. Recently, attempts to establish criteria to identify events that are "hemichannel" mediated and those to allow the distinction between connexin- from pannexin-mediated events have been proposed. Here, I present an update on that topic and discuss the most recent findings related to the nature of functional "hemichannels" focusing on connexin43 and pannexin1. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.
Collapse
Affiliation(s)
- Eliana Scemes
- Dominick P.Purpura Department of Neuroscience, Albert Einstein Collegeof Medicine, Bronx, NY, USA.
| |
Collapse
|
39
|
Rhett JM, Jourdan J, Gourdie RG. Connexin 43 connexon to gap junction transition is regulated by zonula occludens-1. Mol Biol Cell 2011; 22:1516-28. [PMID: 21411628 PMCID: PMC3084674 DOI: 10.1091/mbc.e10-06-0548] [Citation(s) in RCA: 225] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cx43 gap junctions (GJs) are integral to the function of the mammalian heart. It is shown that ZO-1 dynamically regulates the transition between Cx43 connexons and GJ intercellular channels, determining the balance of connexon-mediated cell permeability to GJ communication. Importantly, a novel domain proximal to GJs is identified—the perinexus. Connexin 43 (Cx43) is a gap junction (GJ) protein widely expressed in mammalian tissues that mediates cell-to-cell coupling. Intercellular channels comprising GJ aggregates form from docking of paired connexons, with one each contributed by apposing cells. Zonula occludens-1 (ZO-1) binds the carboxy terminus of Cx43, and we have previously shown that inhibition of the Cx43/ZO-1 interaction increases GJ size by 48 h. Here we demonstrated that increases in GJ aggregation occur within 2 h (∼Cx43 half-life) following disruption of Cx43/ZO-1. Immunoprecipitation and Duolink protein–protein interaction assays indicated that inhibition targets ZO-1 binding with Cx43 in GJs as well as connexons in an adjacent domain that we term the “perinexus.” Consistent with GJ size increases being matched by decreases in connexons, inhibition of Cx43/ZO-1 reduced the extent of perinexal interaction, increased the proportion of connexons docked in GJs relative to undocked connexons in the plasma membrane, and increased GJ intercellular communication while concomitantly decreasing hemichannel-mediated membrane permeance in contacting, but not noncontacting, cells. ZO-1 small interfering RNA and overexpression experiments verified that loss and gain of ZO-1 function govern the transition of connexons into GJs. It is concluded that ZO-1 regulates the rate of undocked connexon aggregation into GJs, enabling dynamic partitioning of Cx43 channel function between junctional and proximal nonjunctional domains of plasma membrane.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
40
|
Kidder GM, Vanderhyden BC. Bidirectional communication between oocytes and follicle cells: ensuring oocyte developmental competence. Can J Physiol Pharmacol 2010; 88:399-413. [PMID: 20555408 DOI: 10.1139/y10-009] [Citation(s) in RCA: 204] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Female fertility is determined to a large extent by the quality (developmental competence) of the oocyte as reflected in its ability to undergo meiosis, be fertilized, and give rise to a healthy embryo. Growth of the mammalian oocyte is coordinated with that of the follicle that encloses it by the actions of signals that pass in both directions between the germline and somatic components. This review summarizes what is known about the roles played by 2 different modes of intrafollicular signalling in oogenesis: paracrine factors activating receptors on the opposite cell type, and direct sharing of small molecules throughout the follicle via gap junction channels. Recent evidence indicates that these 2 modes of signalling interact to regulate oocyte growth and granulosa cell proliferation and that defects in either can contribute to female infertility.
Collapse
Affiliation(s)
- Gerald M Kidder
- Departments of Physiology and Pharmacology, Obstetrics and Gynaecology, and Paediatrics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada.
| | | |
Collapse
|
41
|
Song D, Liu X, Liu R, Yang L, Zuo J, Liu W. Connexin 43 hemichannel regulates H9c2 cell proliferation by modulating intracellular ATP and [Ca2+]. Acta Biochim Biophys Sin (Shanghai) 2010; 42:472-82. [PMID: 20705586 DOI: 10.1093/abbs/gmq047] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Connexin 43 (Cx43), known to be the main protein building blocks of gap junctions and hemichannels in mammalian heart, plays an important role in cardiocytes proliferation. Gap junctional intercellular communication has been suggested to be necessary for cellular proliferation and differentiation. However, the effect of Cx43 hemichannel on cardiocytes proliferation and the mechanism remain unclear. In this study, rat heart cell line H9c2 was used. The Cx43 location, the proliferation rate and hemichannel activity of H9c2 cells and Wnt-3a(+)-H9c2 cells were investigated and the changes of intracellular ATP and [Ca(2+)] were determined. Results showed that the inhibited hemichannel induced by 18beta-glycyrrhetinic acid (GA) evoked intracellular ATP and [Ca(2+)] increase and enhanced H9c2 cell proliferation. Wnt-3a(+)-H9c2 cells displayed enhanced hemichannel activity and proliferation rate. Inhibited hemichannel of Wnt-3a(+)-H9c2 cells induced by 18beta-GA decreased intracellular ATP, increased [Ca(2+)], and enhanced the proliferation of H9c2 cells. This study validated the role of hemichannel in H9c2 cell proliferation regulation, and showed a mechanism involved in the regulation of H9c2 cell proliferation. The proliferation could be enhanced by Cx43 hemichannel-mediated ATP release accompanying intracellular [Ca(2+)] change. However, different changes of ATP were observed in Wnt-3a(+)-H9c2 cells. These findings provided new insights into the molecular mechanisms of proliferation regulation in H9c2 cells and the effect of Wnt-3a on intracellular ATP.
Collapse
Affiliation(s)
- Dongli Song
- Department of Cellular and Genetic Medicine, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
42
|
Wang HX, Li TY, Kidder GM. WNT2 regulates DNA synthesis in mouse granulosa cells through beta-catenin. Biol Reprod 2010; 82:865-75. [PMID: 20107203 DOI: 10.1095/biolreprod.109.080903] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
WNTs are secreted extracellular signaling molecules that transduce their signals by binding to G protein-coupled receptors of the frizzled (FZD) family. They control diverse developmental processes, such as cell fate specification, cell proliferation, cell differentiation, and apoptosis. Although WNT signaling has been shown to be essential for development of the ovary, its mechanistic role in folliculogenesis within the adult ovary has not been studied extensively. Therefore, the objective of this study was to investigate the regulation and function of WNT2 signaling in mouse granulosa cells. Immunostaining identified WNT2 as being expressed in granulosa cells throughout folliculogenesis, but with varying signal strength: in sequential sections, WNT2 immunoreactivity was strongest in healthy antral follicles but weak in atretic follicles. Knockdown of WNT2 expression using transfected short interfering RNA decreased DNA synthesis in granulosa cells, whereas WNT2 overexpression using a recombinant viral vector enhanced it. WNT2 knockdown led to accumulation of glycogen synthase kinase-3beta (GSK3B) in the cytoplasm but reduced the expression of beta-catenin. Conversely, WNT2 overexpression reduced the expression of GSK3B in the cytoplasm and induced beta-catenin translocation from the membrane into the nucleus. Beta-catenin knockdown also inhibited DNA synthesis in granulosa cells and neutralized the effect of WNT2 overexpression. WNT2/beta-catenin signaling had a slight effect on the apoptosis of granulosa cells. Taken together, the data indicate that WNT2 regulates beta-catenin localization in granulosa cells, and WNT2/beta-catenin signaling contributes to regulating their proliferation.
Collapse
Affiliation(s)
- Hong-Xing Wang
- Departments of Physiology and Pharmacology, Obstetrics and Gynecology, and Pediatrics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
43
|
Schalper KA, Palacios-Prado N, Orellana JA, Sáez JC. Currently Used Methods for Identification and Characterization of Hemichannels. ACTA ACUST UNITED AC 2009; 15:207-18. [DOI: 10.1080/15419060802014198] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
44
|
Bintig W, Baumgart J, Walter WJ, Heisterkamp A, Lubatschowski H, Ngezahayo A. Purinergic signalling in rat GFSHR-17 granulosa cells: an in vitro model of granulosa cells in maturing follicles. J Bioenerg Biomembr 2009; 41:85-94. [PMID: 19191015 DOI: 10.1007/s10863-009-9199-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2008] [Accepted: 01/21/2009] [Indexed: 12/20/2022]
Abstract
Purinergic signalling in rat GFSHR-17 granulosa cells was characterised by Ca(2+)-imaging and perforated patch-clamp. We observed a resting intracellular Ca(2+)-concentration ([Ca(2+)](i)) of 100 nM and a membrane potential of -40 mV. This was consistent with high K(+)- and Cl(-) permeability and a high intracellular Cl(-) concentration of 40 mM. Application of ATP for 5-15 s every 3 min induced repeated [Ca(2+)](i) increases and a 30 mV hyperpolarization. The phospholipase C inhibitor U73122 or the IP(3)-receptor antagonist 2-aminoethoethyl diphenyl borate suppressed ATP responses. Further biochemical and pharmacological experiments revealed that ATP responses were related to stimulation of P2Y(2) and P2Y(4) receptors and that the [Ca(2+)](i) increase was a prerequisite for hyperpolarization. Inhibitors of Ca(2+)-activated channels or K(+) channels did not affect the ATP-evoked responses. Conversely, inhibitors of Cl(-) channels hyperpolarized cells to -70 mV and suppressed further ATP-evoked hyperpolarization. We propose that P2Y(2) and P2Y(4) receptors in granulosa cells modulate Cl(-) permeability by regulating Ca(2+)-release.
Collapse
Affiliation(s)
- Willem Bintig
- Institute of Biophysics, Leibniz University Hannover, Herrenhäuser-Str. 2, D-30419 Hannover, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Scemes E, Spray DC, Meda P. Connexins, pannexins, innexins: novel roles of "hemi-channels". Pflugers Arch 2008; 457:1207-26. [PMID: 18853183 DOI: 10.1007/s00424-008-0591-5] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Accepted: 09/17/2008] [Indexed: 12/11/2022]
Affiliation(s)
- Eliana Scemes
- The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | | | | |
Collapse
|
46
|
ATP release from vascular endothelia occurs across Cx43 hemichannels and is attenuated during hypoxia. PLoS One 2008; 3:e2801. [PMID: 18665255 PMCID: PMC2474679 DOI: 10.1371/journal.pone.0002801] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2008] [Accepted: 07/05/2008] [Indexed: 01/22/2023] Open
Abstract
Background Extracellular ATP is an important signaling molecule for vascular adaptation to limited oxygen availability (hypoxia). Here, we pursued the contribution of vascular endothelia to extracellular ATP release under hypoxic conditions. Methodology, Principal Findings We gained first insight from studying ATP release from endothelia (HMEC-1) pre-exposed to hypoxia. Surprisingly, we found that ATP release was significantly attenuated following hypoxia exposure (2% oxygen, 22±3% after 48 h). In contrast, intracellular ATP was unchanged. Similarly, lactate-dehydrogenase release into the supernatants was similar between normoxic or hypoxic endothelia, suggesting that differences in lytic ATP release between normoxia or hypoxia are minimal. Next, we used pharmacological strategies to study potential mechanisms for endothelial-dependent ATP release (eg, verapamil, dipyridamole, 18-alpha-glycyrrhetinic acid, anandamide, connexin-mimetic peptides). These studies revealed that endothelial ATP release occurs – at least in part - through connexin 43 (Cx43) hemichannels. A real-time RT-PCR screen of endothelial connexin expression showed selective repression of Cx43 transcript and additional studies confirmed time-dependent Cx43 mRNA, total and surface protein repression during hypoxia. In addition, hypoxia resulted in Cx43-serine368 phosphorylation, which is known to switch Cx43 hemi-channels from an open to a closed state. Conclusions/Significance Taken together, these studies implicate endothelial Cx43 in hypoxia-associated repression of endothelial ATP release.
Collapse
|
47
|
Krysko DV, Diez-Fraile A, Criel G, Svistunov AA, Vandenabeele P, D’Herde K. Life and death of female gametes during oogenesis and folliculogenesis. Apoptosis 2008; 13:1065-87. [DOI: 10.1007/s10495-008-0238-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Accepted: 06/26/2008] [Indexed: 12/27/2022]
|
48
|
Evans WH, Leybaert L. Mimetic peptides as blockers of connexin channel-facilitated intercellular communication. ACTA ACUST UNITED AC 2008; 14:265-73. [PMID: 18392994 DOI: 10.1080/15419060801891034] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
There is a dearth of chemical inhibitors of connexin-mediated intercellular communication. The advent of short "designer" connexin mimetic peptides has provided new tools to inhibit connexin channels quickly and reversibly. This perspective describes the development of mimetic peptides, especially Gap 26 and 27 that are the most popular and correspond to specific sequences in the extracellular loops of connexins 37, 40 and 43. Initially they were used to inhibit gap-junctional coupling in a wide range of mammalian cells and tissues. Currently, they are also being examined as therapeutic agents that accelerate wound healing and in the early treatment of spinal cord injury. The mimetic peptides bind to connexin hemichannels, influencing channel properties as shown by lowering of electrical conductivity and potently blocking the entry of small reporter dyes and the release of ATP by cells. A mechanism is proposed to help explain the dual action of connexin mimetic peptides on connexin hemichannels and gap-junctional coupling.
Collapse
Affiliation(s)
- W Howard Evans
- Department of Medical Biochemistry and Immunology, Wales Heart Research Institute Cardiff University Medical School, Heath Park, Cardiff, UK.
| | | |
Collapse
|